Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
J Immunother Cancer ; 12(6)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926151

ABSTRACT

BACKGROUND: Lung metastasis is the primary cause of breast cancer-related mortality. Neutrophil extracellular traps (NETs) are involved in the progression of breast cancer. However, the mechanism of NET formation is not fully understood. This study posits that tumor cell-released autophagosomes (TRAPs) play a crucial role in this process. METHODS: TRAPs were isolated from breast cancer cell lines to analyze their impact on NET formation in both human and mouse neutrophils. The study used both in vitro and in vivo models, including Toll-like receptor 4 (TLR4-/-) mice and engineered breast cancer cell lines. Immunofluorescence, ELISA, Western blotting, RNA sequencing, and flow cytometry were employed to dissect the signaling pathways leading to NET production and to explore their immunosuppressive effects, particularly focusing on the impact of NETs on T-cell function. The therapeutic potential of targeting TRAP-induced NETs and their immunosuppressive functions was evaluated using DNase I and αPD-L1 antibodies. Clinical relevance was assessed by correlating circulating levels of TRAPs and NETs with lung metastasis in patients with breast cancer. RESULTS: This study showed that TRAPs induced the formation of NETs in both human and mouse neutrophils by using the high mobility group box 1 and activating the TLR4-Myd88-ERK/p38 signaling axis. More importantly, PD-L1 carried by TRAP-induced NETs inhibited T-cell function in vitro and in vivo, thereby contributing to the formation of lung premetastatic niche (PMN) immunosuppression. In contrast, Becn1 KD-4T1 breast tumors with decreased circulating TRAPs in vivo reduced the formation of NETs, which in turn attenuated the immunosuppressive effects in PMN and resulted in a reduction of breast cancer pulmonary metastasis in murine models. Moreover, treatment with αPD-L1 in combination with DNase I that degraded NETs restored T-cell function and significantly reduced tumor metastasis. TRAP levels in the peripheral blood positively correlated with NET levels and lung metastasis in patients with breast cancer. CONCLUSIONS: Our results demonstrate a novel role of TRAPs in the formation of PD-L1-decorated NETs, which may provide a new strategy for early detection and treatment of pulmonary metastasis in patients with breast cancer.


Subject(s)
Autophagosomes , B7-H1 Antigen , Breast Neoplasms , Extracellular Traps , Lung Neoplasms , Animals , Humans , Mice , Female , Breast Neoplasms/pathology , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Lung Neoplasms/secondary , Extracellular Traps/metabolism , B7-H1 Antigen/metabolism , Autophagosomes/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Line, Tumor
2.
Clin Cancer Res ; 27(23): 6543-6558, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34426437

ABSTRACT

PURPOSE: There is still no standard nonsurgical regimen for conventional chondrosarcoma (CHS). We aimed to identify whether any CHSs have a favored microenvironment for immunotherapy via multidimensional evaluation of the immunologic characteristics of this tumor. EXPERIMENTAL DESIGN: We obtained 98 newly-diagnosed CHS fresh tumors from several institutions and performed comprehensive analysis of data from CyTOF, whole-exome sequencing, and flow cytometry in 22 cases. Clinical data from immunotherapy responders and nonresponders were compared to explore possible biomarkers of immunotherapy response. Mechanism studies were conducted to interpret the biomarker phenotype. RESULTS: Based on the integrated data of single-cell CyTOF and flow cytometry, the CHS immune-microenvironment phenotypes were classified into three groups: subtype I, the "granulocytic-myeloid-derived suppressor cell (G-MDSC) dominant" cluster, with high number of HLA-DR- CD14- myeloid cells; subtype II, the "immune exhausted" cluster, with high exhausted T-cell and dendritic-cell infiltration; and subtype III, the "immune desert" cluster, with few immune cells. Immune cell-rich subtypes (subtype I and II) were characterized by IDH mutation, pathologic high grade, and peritumoral edema, while subtype I cases were exclusively featured by myxoid transformation. In clinical practice involving 12 individuals who received PD-1 antibody immunotherapy, all of the 3 cases with controlled diseases were retrospectively classified as subtype II. In mechanism, IDH mutation significantly elevated chemokine levels and immune-cell infiltration in immune-inactivated tumors. CONCLUSIONS: This study is the first to provide immune characterization of CHS, representing a major step to precise immunotherapy against this malignancy. Immunotherapy is promising for the "immune exhausted" subtype of patients with CHS.


Subject(s)
Chondrosarcoma , Myeloid-Derived Suppressor Cells , Chondrosarcoma/genetics , Chondrosarcoma/therapy , Humans , Immunotherapy/methods , Retrospective Studies , Tumor Microenvironment/genetics
3.
Br J Neurosurg ; : 1-7, 2021 Feb 04.
Article in English | MEDLINE | ID: mdl-33538210

ABSTRACT

BACKGROUND: The selection of a correct level in lumbar spinal stenosis (LSS) remains a common problem and is critically important to the effectiveness of this surgical treatment. Surgery is invasive, and extended laminectomy may lead to secondary surgical complications. The application of diffuse tensor imagining (DTI) and paraspinal mapping (PM) in addition to conventional magnetic resonance imaging (cMRI) may be helpful in this respect. However, the superiority of cMRI + DTI over cMRI+ (DTI or PM) in reducing decompression has not yet been established. METHODS: We compared the surgical levels, determined by cMRI + DTI and cMRI+ (DTI or PM) (self-control). Treatment outcome measurements were performed at two weeks, three months, six months, and twelve months postoperatively. RESULTS: The surgical levels determined by cMRI ± DTI showed less than that determined by cMRI± (DTI or PM) with statistically significant differences (p value = 0.0199) and cMRI ± PM with no statistically significant differences (p value = 0.5503). CONCLUSIONS: The effectiveness of cMRI ± DTI in the reduction of the surgical levels in degenerative lumbar spinal stenosis is superior than that of cMRI± (DTI or PM).

4.
Eur J Nucl Med Mol Imaging ; 48(1): 53-66, 2021 01.
Article in English | MEDLINE | ID: mdl-32592040

ABSTRACT

PURPOSE: Castration-resistant prostate cancer (CRPC) is the most common cause of death in men. The effectiveness of HDAC inhibitors has been demonstrated by preclinical models, but not in clinical studies, probably due to the ineffectively accumulation of HDACI in prostate cancer cells. The purpose of this work was to evaluate effects of a novel HDACI (CN133) on CRPC xenograft model and 22Rv1 cells, and develops methods, PET/CT imaging, to detect the therapeutic effects of CN133 on this cancer. METHODS: We designed and performed study to compare the effects of CN133 with SAHA on the 22Rv1 xenograft model and 22Rv1 cells. Using PET/CT imaging with [11C] Martinostat and [18F] FDG, we imaged mice bearing 22Rv1 xenografts before and after 21-day treatment with placebo and CN133 (1 mg/kg), and uptake on pre-treatment and post-treatment imaging was measured. The anti-tumor mechanisms of CN133 were investigated by qPCR, western blot, and ChIP-qPCR. RESULTS: Our data showed that the CN133 treatment led to a 50% reduction of tumor volume compared to the placebo that was more efficacious than SAHA treatment in this preclinical model. [11C] Martinostat PET imaging could identify early lesions of prostate cancer and can also be used to monitor the therapeutic effect of CN133 in CRPC. Using pharmacological approaches, we demonstrated that effects of CN133 showed almost 100-fold efficacy than SAHA treatment in the experiment of cell proliferation, invasion, and migration. The anti-tumor mechanisms of CN133 were due to the inhibition of AR signaling pathway activity by decreased HDAC 2 and 3 protein expressions. CONCLUSION: Taken together, these studies provide not only a novel epigenetic approach for prostate cancer therapy but also offering a potential tool, [11C] Martinostat PET/CT imaging, to detect the early phase of prostate cancer and monitor therapeutic effect of CN133. These results will likely lead to human trials in the future.


Subject(s)
Histone Deacetylase Inhibitors , Prostatic Neoplasms, Castration-Resistant , Animals , Cell Line, Tumor , Cell Proliferation , Histone Deacetylase Inhibitors/therapeutic use , Humans , Male , Mice , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms, Castration-Resistant/diagnostic imaging , Prostatic Neoplasms, Castration-Resistant/drug therapy , Xenograft Model Antitumor Assays
5.
J Int Med Res ; 48(12): 300060520950934, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33349106

ABSTRACT

BACKGROUND: Femoral head collapse and coxa vara lead to internal fixator failure in elderly patients with hip fracture. External fixator application is an optimal choice; however, the existing methods have many disadvantages. METHODS: Type 31-A1.3 hip fracture models were developed in nine pairs of 1-year-old fresh bovine corpse femur specimens. Each left femur specimen was fixed by a dynamic hip screw (control group), and each right femur specimen was fixed by the slide-poking external fixator (experimental group). Vertical loading and torsion tests were then performed in both groups. RESULTS: In the vertical loading experiment, a 1000-N load was implemented. The mean vertical downward displacement of the femoral head in the experimental and control groups was 1.49322 ± 0.116280 and 2.13656 ± 0.166374 mm, respectively. In the torsion experiment, when the torsion was increased to 10.0 Nm, the mean torsion angle in the experimental and control groups was 7.9733° ± 1.65704° and 15.4889° ± 0.73228°, respectively. The slide-poking external fixator was significantly more resistant to compression and rotation than the dynamic hip screw. CONCLUSION: The slide-poking external fixator for hip fractures that was designed and developed in this study can provide sufficient stability to resist compression and rotation in hip fractures.


Subject(s)
External Fixators , Hip Fractures , Aged , Animals , Biomechanical Phenomena , Bone Screws , Cattle , Fracture Fixation, Internal , Hip Fractures/surgery , Humans , Infant , Internal Fixators
6.
Cell Signal ; 76: 109800, 2020 12.
Article in English | MEDLINE | ID: mdl-33011290

ABSTRACT

NK1.1- CD4+ NKG2D+ T cells are a subpopulation of regulatory T cells that downregulate the functions of CD4+ T, CD8+ T, natural killer (NK) cells, and macrophages through TGF-ß1 production. Early growth response genes 2 (Egr2) and 3 (Egr3) maintain immune homeostasis by modulating T lymphocyte development, inhibiting effector T cell function, and promoting the induction of regulatory T cells. Whether Egr2 and Egr3 directly regulate TGF-ß1 transcription in NK1.1- CD4+ NKG2D+ T cells remains elusive. The expression levels of Egr2 and Egr3 were higher in NK1.1- CD4+ NKG2D+ T cells than in NK1.1- CD4+ NKG2D- T cells. Egr2 and Egr3 expression were remarkably increased after stimulating NK1.1- CD4+ NKG2D+ T cells with sRAE or α-CD3/sRAE. The ectopic expression of Egr2 or Egr3 resulted in the enhancement of TGF-ß1 expression, while knockdown of Egr2 or Egr3 led to the decreased expression of TGF-ß1 in NK1.1- CD4+ NKG2D+ T cells. Egr2 and Egr3 directly bound with the TGF-ß1 promoter as demonstrated by the electrophoretic mobility shift assay and dual-luciferase gene reporter assay. Furthermore, the Egr2 and Egr3 expression of NK1.1- CD4+ NKG2D+ T cells could be induced by the AP-1 and NF-κB transcriptional factors, but had no involvement with the activation of NF-AT and STAT3. In conclusion, Egr2 and Egr3 induced by AP-1 and NF-κB directly initiate TGF-ß1 transcription in NK1.1- CD4+ NKG2D+ T cells. This study indicates that manipulating Egr2 and Egr3 expression would potentiate or alleviate the regulatory function of NK1.1- CD4+ NKG2D+ T cells and this strategy could be used in the therapy for patients with autoimmune diseases or tumor.


Subject(s)
CD4-Positive T-Lymphocytes , Early Growth Response Protein 2/immunology , Early Growth Response Protein 3/immunology , Transforming Growth Factor beta1/immunology , Animals , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Mice , Mice, Transgenic , NF-kappa B/immunology , Neoplasms/immunology
7.
Anal Cell Pathol (Amst) ; 2020: 8546231, 2020.
Article in English | MEDLINE | ID: mdl-32908815

ABSTRACT

Platelet-rich plasma (PRP) and its byproduct platelet-poor plasma (PPP) are rich sources of cytokines in tissue damage repair. Bone marrow-derived mesenchymal stem cells (BM-MSCs) have received more and more attention for their ability to treat multiple diseases. The purpose of our study was to investigate the biologic action of PPP and PRP on BM-MSCs. The adipogenic potential of BM-MSCs revealed no obvious change, but the osteogenic ability of BM-MSCs was enhanced after treated with PRP. CCK8 assays and cell colony formation assays showed that PRP promoted cell proliferation, while this effect of PPP was not obvious. No obvious difference was found in cell cycle and apoptosis of BM-MSCs between PRP and PPP treatment. Expression of ß-galactosidase, a biological marker of senescence, was decreased upon PRP treatment which indicated that PRP provided significant protection against cellular senescence. The migratory capacity of BM-MSCs was detected by scratch and transwell assays. The results indicated that PRP could affect the migration ability of BM-MSCs. From immunofluorescence detection and western blot, we demonstrated that the level of epithelial-mesenchymal transition-related proteins was changed and several pluripotency marker genes, including Sox2, Sall4, Oct4, and Nanog, were increased. Finally, the expression of the key signal pathway such as PI3K/AKT was examined. Our findings suggested that PRP promoted cell migration of BM-MSCs via stimulating the signaling pathway of PI3K/AKT.


Subject(s)
Mesenchymal Stem Cells/metabolism , Platelet-Rich Plasma/metabolism , Animals , Apoptosis , Cell Cycle , Cell Differentiation , Cell Movement , Cell Proliferation , Cellular Senescence , Male , Mesenchymal Stem Cells/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction
8.
Pharmacol Res ; 159: 105010, 2020 09.
Article in English | MEDLINE | ID: mdl-32544428

ABSTRACT

AMD3100 (plerixafor), a CXCR4 antagonist, has opened a variety of avenues for potential therapeutic approaches in different refractory diseases. The CXCL12/CXCR4 axis and its signaling pathways are involved in diverse disorders including HIV-1 infection, tumor development, non-Hodgkin lymphoma, multiple myeloma, WHIM Syndrome, and so on. The mechanisms of action of AMD3100 may relate to mobilizing hematopoietic stem cells, blocking infection of X4 HIV-1, increasing circulating neutrophils, lymphocytes and monocytes, reducing myeloid-derived suppressor cells, and enhancing cytotoxic T-cell infiltration in tumors. Here, we first revisit the pharmacological discovery of AMD3100. We then review monotherapy of AMD3100 and combination use of AMD3100 with other agents in various diseases. Among those, we highlight the perspective of AMD3100 as an immunomodulator to regulate immune responses particularly in the tumor microenvironment and synergize with other therapeutics. All the pre-clinical studies support the clinical testing of the monotherapy and combination therapies with AMD3100 and further development for use in humans.


Subject(s)
Anti-HIV Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Benzylamines/therapeutic use , Cyclams/therapeutic use , HIV Infections/drug therapy , Neoplasms/drug therapy , Receptors, CXCR4/antagonists & inhibitors , Animals , Anti-HIV Agents/adverse effects , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Benzylamines/adverse effects , Cyclams/adverse effects , Drug Contamination , HIV Infections/immunology , HIV Infections/metabolism , HIV Infections/virology , Humans , Neoplasms/immunology , Neoplasms/metabolism , Primary Immunodeficiency Diseases/drug therapy , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/metabolism , Receptors, CXCR4/metabolism , Signal Transduction , Tumor Microenvironment , Warts/drug therapy , Warts/immunology , Warts/metabolism
9.
Pharmacol Res ; 157: 104843, 2020 07.
Article in English | MEDLINE | ID: mdl-32360582

ABSTRACT

Both antitumor and protumor property of mesenchymal stem cells (MSCs) have been demonstrated. We hypothesize that this contradiction is due to the heterogeneity of MSC subsets and that extracellular vesicles (EVs) from distinct MSC subsets can transfer the corresponding antitumor activities. Here we evaluated the antitumor activities of two subsets of adipose-derived mesenchymal stem cells (ADSCs) and ADSC-derived EVs (ADSC-EVs) in immunocompetent syngeneic mouse models of breast cancer. We identified CD90high and CD90low ADSC subsets and demonstrated that CD90high ADSCs could be converted into CD90low ADSCs by stimulation with LPS. CD90low ADSCs and its derived EVs significantly inhibited tumor growth in tumor-bearing mice. Benefit of tumor control were associated with decreased tumor cell proliferation and migration, and enhanced tumor cell apoptosis mediated by ADSC-EVs. Antioncogenic miRNA-16-5p loaded CD90low ADSC-EVs further significantly enhanced antitumor activities. Taken together, this study represents the first attempt to apply our newly identified antitumor ADSCs and its derived EVs in preclinical treatment of breast cancer. This study also provides the evidence that EVs can serve as a novel and effective therapeutics or drug delivery vesicle. This new therapeutic approach could be potentially applicable to breast cancer and many other types of cancer.


Subject(s)
Breast Neoplasms/therapy , Extracellular Vesicles/transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Adipose Tissue/cytology , Animals , Apoptosis , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Female , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness , Phenotype , Thy-1 Antigens/metabolism , Tumor Burden
10.
Oncotarget ; 10(43): 4479-4491, 2019 Jul 09.
Article in English | MEDLINE | ID: mdl-31320999

ABSTRACT

Both anti-tumoral and pro-tumoral effects of mesenchymal stem cells (MSCs) in preclinical treatment of ovarian cancer have been controversially demonstrated. In this study, we profiled the phenotypes of mouse compact bone-derived MSCs (CB-MSCs) and bone marrow-derived MSCs (BM-MSCs) and found that CB-MSCs expressed lower CD90 compared to BM-MSCs. We examined gene expression of immune regulating cytokines of CB-MSCs in 2D and 3D culture and under stimulation with TLR4 agonist LPS or immune activator VIC-008. Our data showed that when CB-MSCs were cultured in simulated in vivo 3D condition, CD90 expression was further decreased. Moreover, gene expressions of immune activating cytokines IL-12, IL-21, IFNγ and a pro-inflammatory cytokine CXCL10 in CB-MSCs were increased in 3D culture whereas gene expression of anti-inflammatory cytokines IL-10 and CCL5 were downregulated. Stimulation of CB-MSCs by LPS or VIC-008 presented similar profile of the cytokine gene expressions to that in 3D culture which might benefit the anti-tumor efficacy of CD90low MSCs. The anti-tumor effects of CD90low CB-MSCs alone or in combination with VIC-008 were evaluated in a syngeneic orthotopic mouse model of ovarian cancer. Treatment that combines CB-MSCs and VIC-008 significantly decreased tumor growth and prolonged mouse survival. This was associated with the increase of activated anti-tumoral CD4+ and CD8+ T cells and the decrease of Treg cells in the tumor microenvironment. Taken together, our study demonstrates the synergistic anti-tumoral efficacy by application of CB-MSCs combined with immune activator VIC-008 and provides new insight into CD90low MSCs as a new anti-tumor arsenal.

11.
Int J Oncol ; 54(5): 1843-1852, 2019 May.
Article in English | MEDLINE | ID: mdl-30864702

ABSTRACT

Mesenchymal stem cells (MSCs) have been demonstrated to be involved in tumor progression and the modulation of the tumor microenvironment, partly through their secretome. Extracellular vesicles (EVs) are membranous nanovesicles secreted by multiple types of cells and have been demonstrated to mediate intercellular communication in both physiological and pathological conditions. However, numerous questions still remain regarding the underlying mechanisms and functional consequences of these interactions. The purpose of this study was to investigate the effects of human umbilical cord mesenchymal stem cell­derived EVs (hUC­MSC­EVs) on the proliferation, migration and invasion of human breast cancer cells. We successfully generated and identified hUC­MSCs and hUC­MSC­EVs which were used in this study. The results revealed that treatment of the MDA­MB­231 and MCF­7 human breast cancer cells with medium containing hUC­MSC­EVs significantly enhanced the proliferation, migration and invasion of the cells in vitro. Treatment of the cells with medium containing hUC­MSC­EVs also reduced E­cadherin expression and increased N­cadherin expression, thus promoting the epithelial­mesenchymal transition (EMT) of the breast cancer cells. Treatment of the breast cancer cells with extracellular signal­regulated kinase (ERK) inhibitor prior to the interaction with hUC­MSC­EVs significantly reversed the enhanced proliferation, migration and invasion, as well as the EMT of the breast cancer cells induced by the hUC­MSC­EVs. On the whole, these data indicate that hUC­MSC­EVs promote the invasive and migratory potential of breast cancer cells through the induction of EMT via the ERK pathway, leading to malignant tumor progression and metastasis. Taken together, the findings of this study suggest that targeting pathways to reverse EMT may lead to the development of novel therapeutic approaches with which to combat breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Culture Media/pharmacology , Extracellular Vesicles/pathology , MAP Kinase Signaling System , Mesenchymal Stem Cells/cytology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Culture Media/chemistry , Epithelial-Mesenchymal Transition , Extracellular Vesicles/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Mesenchymal Stem Cells/metabolism , Tumor Microenvironment , Umbilical Cord/cytology , Umbilical Cord/metabolism
12.
FASEB J ; 33(5): 6596-6608, 2019 05.
Article in English | MEDLINE | ID: mdl-30802149

ABSTRACT

Blockade of immune-checkpoint programmed cell death protein 1 (PD-1) or programmed cell death ligand 1 can enhance effector T-cell responses. However, the lack of response in many patients to checkpoint-inhibitor therapies emphasizes the need for combination immunotherapies to pursue maximal antitumor efficacy. We have previously demonstrated that antagonism of C-X-C chemokine receptor type 4 (CXCR4) by plerixafor (AMD3100) can decrease regulatory T (Treg)-cell intratumoral infiltration. Therefore, a combination of these 2 therapies might increase antitumor effects. Here, we evaluated the antitumor efficacy of AMD3100 and anti-PD-1 (αPD-1) antibody alone or in combination in an immunocompetent syngeneic mouse model of ovarian cancer. We found that AMD3100, a highly specific CXCR4 antagonist, directly down-regulated the expression of both C-X-C motif chemokine 12 (CXCL12) and CXCR4 in vitro and in vivo in tumor cells. AMD3100 and αPD-1 significantly inhibited tumor growth and prolonged the survival of tumor-bearing mice when given as monotherapy. Combination of these 2 agents significantly enhanced antitumor effects compared with single-agent administration. Benefits of tumor control and animal survival were associated with immunomodulation mediated by these 2 agents, which were characterized by increased effector T-cell infiltration, increased effector T-cell function, and increased memory T cells in tumor microenvironment. Intratumoral Treg cells were decreased, and conversion of Treg cells into T helper cells was increased by AMD3100 treatment. Intratumoral myeloid-derived suppressor cells were decreased by the combined treatment, which was associated with decreased IL-10 and IL-6 in the ascites. Also, the combination therapy decreased suppressive leukocytes and facilitated M2-to-M1 macrophage polarization in the tumor. These results suggest that AMD3100 could be used to target the CXCR4-CXCL12 axis to inhibit tumor growth and prevent multifaceted immunosuppression alone or in combination with αPD-1 in ovarian cancer, which could be clinically relevant to patients with this disease.-Zeng, Y., Li, B., Liang, Y., Reeves, P. M., Qu, X., Ran, C., Liu, Q., Callahan, M. V., Sluder, A. E., Gelfand, J. A., Chen, H., Poznansky, M. C. Dual blockade of CXCL12-CXCR4 and PD-1-PD-L1 pathways prolongs survival of ovarian tumor-bearing mice by prevention of immunosuppression in the tumor microenvironment.


Subject(s)
B7-H1 Antigen , Chemokine CXCL12 , Heterocyclic Compounds/pharmacology , Immune Tolerance/drug effects , Neoplasm Proteins , Ovarian Neoplasms , Programmed Cell Death 1 Receptor , Receptors, CXCR4 , Signal Transduction , Tumor Microenvironment , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Benzylamines , Cell Line, Tumor , Chemokine CXCL12/antagonists & inhibitors , Chemokine CXCL12/immunology , Cyclams , Female , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
13.
J Orthop Surg Res ; 14(1): 23, 2019 Jan 22.
Article in English | MEDLINE | ID: mdl-30670090

ABSTRACT

BACKGROUND: In lumbar spinal stenosis (LSS), at most times, several levels are impaired and selecting the correct level remains a common problem for surgeons, as surgery remains invasive, and extended laminectomy may lead to secondary surgical complications. Therefore, helping to select the correct level may be useful for surgeons. The use of diffuse tensor imaging (DTI) and paraspinal mapping (PM) in addition to conventional magnetic resonance imaging (MRI) may be helpful (Chen et al., J Orthop Surg Res 11:47, 2016). However, with decompression levels determined by conventional magnetic resonance imaging (MRI) increasing, whether the benefits of reducing decompression level of conventional MRI + (DTI or PM) will be more obvious is unknown. METHODS: Reduced surgical levels that were different between levels determined by conventional MRI + (DTI or PM) and conventional MRI + neurogenic examination (NE) between groups were compared. Treatment outcome measures were performed at 2 weeks, 3 months, 6 months, and 12 months postoperatively. RESULTS: The reduced levels of three groups showed no statistically significant differences between each other except for two levels and four levels (two levels/three levels, p = 0.085; two levels/four levels, p = 0.039; three levels/ four levels, p = 0.506, respectively). CONCLUSIONS: With surgical levels determined by conventional MRI increasing, the benefits of DTI and PM will be uncertainly more obvious.


Subject(s)
Decompression, Surgical/trends , Diffusion Tensor Imaging/trends , Lumbar Vertebrae/diagnostic imaging , Paraspinal Muscles/diagnostic imaging , Spinal Stenosis/diagnostic imaging , Adult , Aged , Aged, 80 and over , Decompression, Surgical/methods , Diffusion Tensor Imaging/methods , Female , Follow-Up Studies , Humans , Lumbar Vertebrae/surgery , Magnetic Resonance Imaging/methods , Magnetic Resonance Imaging/trends , Male , Middle Aged , Paraspinal Muscles/surgery , Spinal Stenosis/surgery
14.
Cancer Res ; 78(14): 3938-3953, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29735547

ABSTRACT

The mechanism by which osteosarcomas metastasize is elusive, and challenges remain regarding its treatment with modalities including immunotherapy. CXCL12 is deeply involved in the process of tumor metastasis and T-cell homing, which is driven by a chemokine gradient, but healthy bones are supposed to preferentially express CXCL12. Here, we show for the first time that osteosarcomas epigenetically downregulate CXCL12 expression via DNA methyltransferase 1 (DNMT1) and consequently acquire the ability to metastasize and to impair cytotoxic T-cell homing to the tumor site. Analysis of human osteosarcoma cases further revealed that CXCL12 expression strongly correlated with overall survival. Evaluations on fresh human chemotherapy-free osteosarcoma samples also showed a positive correlation between CXCL12 concentration and the number of intratumoral lymphocytes. Critically, treatment targeting DNMT1 in immunocompetent mouse models significantly elevated expression of CXCL12 in tumors, resulting in a robust immune response and consequently eradicating early lung metastases in addition to suppressing subcutaneous tumor growth. These antitumor effects were abrogated by CXCL12-CXCR4 blockade or CD8+ T-cell depletion. Collectively, our data show that CXCL12 regulation plays a significant role in both tumor progression and immune response, and targeting CXCL12 is promising for therapeutics against osteosarcoma.Significance: Epigenetic regulation of CXCL12 controls metastasis and immune response in osteosarcoma, suggesting epigenetic therapies or therapies targeting CXCL12 have potential for therapeutic intervention in osteosarcoma. Cancer Res; 78(14); 3938-53. ©2018 AACR.


Subject(s)
Chemokine CXCL12/genetics , Epigenesis, Genetic/genetics , Osteosarcoma/genetics , Adolescent , Adult , Aged , Animals , CD8-Positive T-Lymphocytes/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Child , Child, Preschool , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Humans , Lung Neoplasms/genetics , Mice , Middle Aged , Signal Transduction/genetics , T-Lymphocytes, Cytotoxic/physiology , Young Adult
15.
Cancer Immunol Res ; 6(5): 539-551, 2018 05.
Article in English | MEDLINE | ID: mdl-29511032

ABSTRACT

AMD3100 (plerixafor), a CXCR4 antagonist, has been demonstrated to suppress tumor growth and modulate intratumoral T-cell trafficking. However, the effect of AMD3100 on immunomodulation remains elusive. Here, we explored immunomodulation and antitumor efficacy of AMD3100 in combination with a previously developed mesothelin-targeted, immune-activating fusion protein, VIC-008, in two syngeneic, orthotopic models of malignant mesothelioma in immunocompetent mice. We showed that combination therapy significantly suppressed tumor growth and prolonged animal survival in two mouse models. Tumor control and survival benefit were associated with enhanced antitumor immunity. VIC-008 augmented mesothelin-specific CD8+ T-cell responses in the spleen and lymph nodes and facilitated intratumoral lymphocytic infiltration. However, VIC-008 treatment was associated with increased programmed cell death protein-1 (PD-1) expression on intratumoral CD8+ T cells, likely due to high CXCL12 in the tumor microenvironment. AMD3100 alone and in combination with VIC-008 modulated immunosuppression in tumors and the immune system through suppression of PD-1 expression on CD8+ T cells and conversion of regulatory T cells (Tregs) into CD4+CD25-Foxp3+IL2+CD40L+ helper-like cells. In mechanistic studies, we demonstrated that AMD3100-driven Treg reprogramming required T cell receptor (TCR) activation and was associated with loss of PTEN due to oxidative inactivation. The combination of VIC-008 augmentation of tumor-specific CD8+ T-cell responses with AMD3100 abrogation of immunosuppression conferred significant benefits for tumor control and animal survival. These data provide new mechanistic insight into AMD3100-mediated immunomodulation and highlight the enhanced antitumor effect of AMD3100 in combination with a tumor antigen-targeted therapy in mouse malignant mesothelioma, which could be clinically relevant to patients with this difficult-to-treat disease. Cancer Immunol Res; 6(5); 539-51. ©2018 AACR.


Subject(s)
Antigens, Bacterial/immunology , Cancer Vaccines/therapeutic use , GPI-Linked Proteins/immunology , HSP70 Heat-Shock Proteins/immunology , Heterocyclic Compounds/pharmacology , Immunomodulation/drug effects , Mesothelioma/therapy , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/therapeutic use , Benzylamines , CHO Cells , Cancer Vaccines/immunology , Cell Line, Tumor , Combined Modality Therapy , Cricetinae , Cricetulus , Cyclams , Drug Synergism , Female , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/therapeutic use , Heterocyclic Compounds/administration & dosage , Mesothelin , Mesothelioma/immunology , Mesothelioma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use
16.
Proc Natl Acad Sci U S A ; 115(8): 1877-1882, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29437954

ABSTRACT

HIV controllers (HCs) are individuals who can naturally control HIV infection, partially due to potent HIV-specific CD8+ T cell responses. Here, we examined the hypothesis that superior function of CD8+ T cells from HCs is encoded by their T cell receptors (TCRs). We compared the functional properties of immunodominant HIV-specific TCRs obtained from HLA-B*2705 HCs and chronic progressors (CPs) following expression in primary T cells. T cells transduced with TCRs from HCs and CPs showed equivalent induction of epitope-specific cytotoxicity, cytokine secretion, and antigen-binding properties. Transduced T cells comparably, albeit modestly, also suppressed HIV infection in vitro and in humanized mice. We also performed extensive molecular dynamics simulations that provided a structural basis for similarities in cytotoxicity and epitope cross-reactivity. These results demonstrate that the differential abilities of HIV-specific CD8+ T cells from HCs and CPs are not genetically encoded in the TCRs alone and must depend on additional factors.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Epitopes, T-Lymphocyte/genetics , HIV Infections/immunology , HIV-1/immunology , Receptors, Antigen, T-Cell/genetics , Cloning, Molecular , Gene Expression Regulation/immunology , HEK293 Cells , HLA-B27 Antigen , Humans , Jurkat Cells
17.
Mol Med Rep ; 17(4): 5376-5383, 2018 04.
Article in English | MEDLINE | ID: mdl-29393352

ABSTRACT

In recent years, stem cell research has continued to benefit from its crossover with chemistry, particularly the investigation of small molecular drugs modulating specific targets to regulate stem cell fate. Idebenone (IDB) is a yellow crystalline powder that is used in the treatment of chronic cerebrovascular diseases. The objective of the present study was to examine whether IDB had an influence on bone marrow­derived mesenchymal stem cells (BMSCs) extracted from the bone marrow of Sprague­Dawley rats. The effects of IDB on cell proliferation, cell cloning and migration were investigated. Cell cycle, apoptosis, DAPI nuclear staining and senescence­associated ß­galactosidase (SA­ß­gal) staining were also examined. The results revealed that IDB at suitable concentrations enhanced cell cloning capacity, promoted the proliferation of BMSCs, delayed cellular senescence, and inhibited cell apoptosis and migration. Western blot analysis indicated that IDB increased the expression of B­cell lymphoma 2 (Bcl­2), signal transducer and activator of transcription­3, Nanog, octamer­binding transcription factor 4, E­cadherin, proliferating cell nuclear antigen, cyclinD1 and cyclinD3, and decreased the expression of Bcl­2­associated X protein, cleaved caspase­3, N­cadherin, vimentin and α­smooth muscle actin. In conclusion, these experiments confirmed that IDB in low doses had no toxic effect and may exert protective effects on BMSCs.


Subject(s)
Mesenchymal Stem Cells/drug effects , Ubiquinone/analogs & derivatives , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Colony-Forming Units Assay , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Rats , Ubiquinone/pharmacology
18.
Oncotarget ; 9(1): 591-606, 2018 Jan 02.
Article in English | MEDLINE | ID: mdl-29416638

ABSTRACT

Cigarette smoking is a well-known risk factor in the development and progression of malignant diseases. Nicotine, the major constituent in cigarette smoke, has also shown negative effects on stem cells. Mesenchymal stem cells (MSCs) have been widely demonstrated to migrate into tumors and play key roles in cancer progression. However, the mechanisms by which nicotine impacts MSCs and tumorigenesis of lung cancer are still undetermined. In this study we investigated the effects of nicotine on human umbilical cord mesenchymal stem cells (hUC-MSCs) and the impacts of nicotine-treated hUC-MSCs on tumor formation and progression. We found that nicotine has a toxic effect on hUC-MSCs and changes the morphology, inhibits proliferation and promotes apoptosis of hUC-MSCs in a dose-dependent manner. Nicotine-treated hUC-MSCs produce higher level of IL-6. Moreover, nicotine promotes migration, stemness and epithelial-mesenchymal transition (EMT) of hUC-MSCs by inhibiting E-cadherin expression and upregulating mesenchymal markers such as N-cadherin and Vimentin, leading to the induction of stem cell markers Sox2, Nanog, Sall4, Oct4 and CD44. Migration and proliferation of non-small cell lung cancer A549 cells and breast cancer MCF-7 cells are promoted after their coculture with nicotine-treated hUC-MSCs in a cell-cell contact-independent manner. Furthermore, nicotine-treated hUC-MSCs promote tumor formation and growth of A549 cells in nude mice. These studies demonstrated that the enhanced stemness and EMT of hUC-MSCs induced by nicotine are critical for the development of tobacco-related cancers.

19.
Cell Mol Immunol ; 15(8): 768-781, 2018 08.
Article in English | MEDLINE | ID: mdl-29375131

ABSTRACT

We previously identified human phosphatidylethanolamine-binding protein 4 (hPEBP4) as an antiapoptotic protein with increased expression levels in breast, ovarian and prostate cancer cells, but low expression levels in normal tissues, which makes hPEBP4 an attractive target for immunotherapy. Here, we developed hPEBP4-derived immunogenic peptides for inducing antigen-specific cytotoxic T lymphocytes (CTLs) targeting breast cancer. A panel of hPEBP4-derived peptides predicted by peptide-MHC-binding algorithms was evaluated to characterize their HLA-A2.1 affinity and immunogenicity. We identified a novel immunogenic peptide, P40-48 (TLFCQGLEV), that was capable of eliciting specific CTL responses in HLA-A2.1/Kb transgenic mice, as well as in peripheral blood lymphocytes from breast cancer patients. Furthermore, amino-acid substitutions in the P40-48 sequence improved its immunogenicity against hPEBP4, a self-antigen, thus circumventing tolerance. We designed peptide analogs by preferred auxiliary HLA-A*0201 anchor residue replacement, which induced CTLs that were crossreactive to the native peptide. Several analogs were able to stably bind to HLA-A*0201 and elicit specific CTL responses better than the native sequence. Importantly, adoptive transfer of CTLs induced by vaccination with two analogs more effectively inhibited tumor growth than the native peptide. These data indicate that peptide analogs with high immunogenicity represent promising candidates for peptide-mediated therapeutic cancer vaccines.


Subject(s)
Breast Neoplasms/immunology , Epitopes/immunology , HLA-A2 Antigen/metabolism , Peptides/immunology , Peptides/metabolism , Phosphatidylethanolamine Binding Protein/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Breast Neoplasms/blood , Breast Neoplasms/therapy , Cancer Vaccines/immunology , Cytotoxicity, Immunologic , Female , HLA-A2 Antigen/immunology , Humans , Immunotherapy , Kaplan-Meier Estimate , MCF-7 Cells , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Peptides/chemical synthesis , Phosphatidylethanolamine Binding Protein/chemistry , Statistics, Nonparametric
20.
J Virol ; 91(22)2017 11 15.
Article in English | MEDLINE | ID: mdl-28878089

ABSTRACT

Immune control of human immunodeficiency virus type 1 (HIV) infection is typically associated with effective Gag-specific CD8+ T-cell responses. We here focus on HLA-B*14, which protects against HIV disease progression, but the immunodominant HLA-B*14-restricted anti-HIV response is Env specific (ERYLKDQQL, HLA-B*14-EL9). A subdominant HLA-B*14-restricted response targets Gag (DRYFKTLRA, HLA-B*14-DA9). Using HLA-B*14/peptide-saporin-conjugated tetramers, we show that HLA-B*14-EL9 is substantially more potent at inhibiting viral replication than HLA-B*14-DA9. HLA-B*14-EL9 also has significantly higher functional avidity (P < 0.0001) and drives stronger selection pressure on the virus than HLA-B*14-DA9. However, these differences were HLA-B*14 subtype specific, applying only to HLA-B*14:02 and not to HLA-B*14:01. Furthermore, the HLA-B*14-associated protection against HIV disease progression is significantly greater for HLA-B*14:02 than for HLA-B*14:01, consistent with the superior antiviral efficacy of the HLA-B*14-EL9 response. Thus, although Gag-specific CD8+ T-cell responses may usually have greater anti-HIV efficacy, factors independent of protein specificity, including functional avidity of individual responses, are also critically important to immune control of HIV.IMPORTANCE In HIV infection, although cytotoxic T lymphocytes (CTL) play a potentially critical role in eradication of viral reservoirs, the features that constitute an effective response remain poorly defined. We focus on HLA-B*14, unique among HLAs associated with control of HIV in that the dominant CTL response is Env specific, not Gag specific. We demonstrate that Env-specific HLA-B*14-restricted activity is substantially more efficacious than the subdominant HLA-B*14-restricted Gag response. Env immunodominance over Gag and strong Env-mediated selection pressure on HIV are observed only in subjects expressing HLA-B*14:02, and not HLA-B*14:01. This reflects the increased functional avidity of the Env response over Gag, substantially more marked for HLA-B*14:02. Finally, we show that HLA-B*14:02 is significantly more strongly associated with viremic control than HLA-B*14:01. These findings indicate that, although Gag-specific CTL may usually have greater anti-HIV efficacy than Env responses, factors independent of protein specificity, including functional avidity, may carry greater weight in mediating effective control of HIV.


Subject(s)
HIV Envelope Protein gp160/immunology , HIV Infections/immunology , HIV-1/immunology , HLA-B14 Antigen/immunology , Immunity, Cellular , Peptides/immunology , gag Gene Products, Human Immunodeficiency Virus/immunology , Adult , CD8-Positive T-Lymphocytes , HIV Infections/pathology , HIV Infections/therapy , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...