Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
Elife ; 122024 May 15.
Article En | MEDLINE | ID: mdl-38747577

Certain bacteria demonstrate the ability to target and colonize the tumor microenvironment, a characteristic that positions them as innovative carriers for delivering various therapeutic agents in cancer therapy. Nevertheless, our understanding of how bacteria adapt their physiological condition to the tumor microenvironment remains elusive. In this work, we employed liquid chromatography-tandem mass spectrometry to examine the proteome of E. coli colonized in murine tumors. Compared to E. coli cultivated in the rich medium, we found that E. coli colonized in tumors notably upregulated the processes related to ferric ions, including the enterobactin biosynthesis and iron homeostasis. This finding indicated that the tumor is an iron-deficient environment to E. coli. We also found that the colonization of E. coli in the tumor led to an increased expression of lipocalin 2 (LCN2), a host protein that can sequester the enterobactin. We therefore engineered E. coli in order to evade the nutritional immunity provided by LCN2. By introducing the IroA cluster, the E. coli synthesizes the glycosylated enterobactin, which creates steric hindrance to avoid the LCN2 sequestration. The IroA-E. coli showed enhanced resistance to LCN2 and significantly improved the anti-tumor activity in mice. Moreover, the mice cured by the IroA-E. coli treatment became resistant to the tumor re-challenge, indicating the establishment of immunological memory. Overall, our study underscores the crucial role of bacteria's ability to acquire ferric ions within the tumor microenvironment for effective cancer therapy.


Escherichia coli , Iron , Lipocalin-2 , Animals , Escherichia coli/genetics , Escherichia coli/metabolism , Lipocalin-2/metabolism , Lipocalin-2/genetics , Mice , Iron/metabolism , Neoplasms/therapy , Neoplasms/immunology , Enterobactin/metabolism , Tumor Microenvironment , Cell Line, Tumor
2.
EMBO Mol Med ; 16(2): 416-428, 2024 Feb.
Article En | MEDLINE | ID: mdl-38225455

The tumor microenvironment (TME) presents differential selective pressure (DSP) that favors the growth of cancer cells, and monovalent therapy is often inadequate in reversing the cancer cell dominance in the TME. In this work, we introduce bacteria as a foreign species to the TME and explore combinatorial treatment strategies to alter DSP for tumor eradication. We show that cancer-selective chemotherapeutic agents and fasting can provide a strong selection pressure against tumor growth in the presence of bacteria. Moreover, we show that an immunogenic drug (oxaliplatin), but not a non-immunogenic one (5-FU), synergizes with the bacteria to activate both the innate and adaptive immunity in the TME, resulting in complete tumor remission and a sustained anti-tumor immunological memory in mice. The combination of oxaliplatin and bacteria greatly enhances the co-stimulatory and antigen-presenting molecules on antigen-presenting cells, which in turn bridge the cytotoxic T cells for cancer-cell killing. Our findings indicate that rational combination of bacterial therapy and immunogenic chemotherapy can promote anticancer immunity against the immunosuppressive TME.


Antineoplastic Agents , Neoplasms , Animals , Mice , Oxaliplatin/therapeutic use , Tumor Microenvironment , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , T-Lymphocytes, Cytotoxic , Immunotherapy/methods , Cell Line, Tumor
3.
ACS Nano ; 17(15): 14532-14544, 2023 08 08.
Article En | MEDLINE | ID: mdl-37466994

Direct delivery of therapeutic genes is a promising approach for treating cancers and other diseases. The current human viral vectors, however, suffer from several drawbacks, including poor cell-type specificity and difficult large-scale production. The M13 phage provides an alternative vehicle for gene therapy with engineerable specificity, but the low transduction efficiency seriously limits its translational application. In this work, we discovered important factors of cells and phages that greatly influence the phage transduction. The up-regulation of PrimPol or the down-regulation of DMBT1 in cells significantly enhanced the phage transduction efficiency. Furthermore, we found that the phage transduction efficiency was inversely correlated with the phage size. By carefully reconstructing the phage origin with the gene of interest, we designed "TransPhage" with a minimal length and maximal transduction efficiency. We showed that TransPhage successfully transduced the human cells with an excellent efficiency (up to 95%) comparable to or superior to that of the adeno-associated virus vectors. Moreover, we showed that TransPhage's tropism was specific to the cells that overexpress the target antigen, whereas adeno-associated viruses (AAVs) promiscuously infected many cell types. Using TransPhage as a gene therapy vehicle, we invented an NK-cell-mediated immunotherapy in which a membrane-bound fragment crystallizable region was introduced to cancer cells. We showed in vitro that the cancer cells expressing the membrane-bound fragment crystallizable (Fc) were effectively killed by CD16+ NK cells through an antibody-dependent cell-mediated cytotoxicity (ADCC)-like mechanism. In the xenograft mouse model, the administration of TransPhage carrying the membrane-bound Fc gene greatly suppressed tumor growth.


Gene Transfer Techniques , Neoplasms , Humans , Mice , Animals , Genetic Vectors , Bacteriophage M13 , Genetic Therapy , Killer Cells, Natural , Neoplasms/genetics , Neoplasms/therapy , Calcium-Binding Proteins , DNA-Binding Proteins , Tumor Suppressor Proteins/genetics , DNA-Directed DNA Polymerase , DNA Primase/genetics , Multifunctional Enzymes
4.
Cell Oncol (Dordr) ; 46(3): 643-660, 2023 Jun.
Article En | MEDLINE | ID: mdl-36607556

BACKGROUND: Reprogramming of metabolism is strongly associated with the development of cancer. However, the role of metabolic reprogramming in the remodeling of pre-metastatic niche (PMN), a key step in metastasis, is still unknown. We aimed to investigate the metabolic alternation during lung PMN formation in breast cancer. METHODS: We assessed the transcriptomes and lipidomics of lung of MMTV-PyVT mice by microarray and liquid chromatography-tandem mass mass spectrometry before lung metastasis. The validation of gene or protein expressions was performed by quantitative real-time polymerase chain reaction or immunoblot and immunohistochemistry respectively. The lung fibroblasts were isolated from mice and then co-cultured with breast cancer to identify the influence of cancer on the change of lung fibroblasts in PMN. RESULTS: We demonstrated changes in the lipid profile and several lipid metabolism genes in the lungs of breast cancer-bearing MMTV-PyVT mice before cancer spreading. The expression of ACACA (acetyl-CoA carboxylase α) was downregulated in the lung fibroblasts, which contributed to changes in acetylation of protein's lysine residues and the synthesis of fatty acid. The downregulation of ACACA in lung fibroblasts triggered a senescent and inflammatory phenotypic shift of lung fibroblasts in both in vivo and in vitro models. The senescence-associated secretory phenotype of lung fibroblasts enabled the recruitment of immunosuppressive granulocytic myeloid-derived suppressor cells into the lungs through the production of CXCL1 in the lungs. Knock-in of ACACA prevented lung metastasis in the MMTV-PyVT mouse model, further supporting that ACACA was involved in the remodeling of the lung PMN. CONCLUSIONS: Taken together, these data revealed a mechanism by which ACACA downregulation directed the formation of an immunosuppressive lung PMN in breast cancer.


Acetyl-CoA Carboxylase , Breast Neoplasms , Cellular Senescence , Fibroblasts , Lung Neoplasms , Animals , Mice , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cellular Senescence/genetics , Down-Regulation , Fibroblasts/metabolism , Fibroblasts/pathology , Lung/metabolism , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Humans
5.
Oncogene ; 39(4): 739-753, 2020 01.
Article En | MEDLINE | ID: mdl-31558801

Metastatic tumors have been shown to establish a supportive pre-metastatic niche (PMN) in distant organs, which in turn determines disseminated tumor cells' targeting of such organs. PMN is formed through the recruitment of bone-marrow-derived cells (BMDCs); however, the role of BMDCs in PMN formation is not fully understood. On the basis of RNA-seq data and bioinformatic analysis, secretion of extracellular vesicle (EV) miR-92a by BMDCs of lung cancer-bearing mice contributes to the establishment of liver PMN. Both BMDC-derived EVs and miR-92a mimics potentiate the activation of hepatic stellate cells (HSCs), subsequently increasing extracellular matrix (ECM) deposition in mice. Consequently, remodeling of the liver microenvironment enhanced immunosuppressive cell accumulation and cancer cell attachment. EVs miR-92a directly suppressed its target SMAD7, leading to the enhancement of transforming growth factor-ß signaling in HSC. Elevated levels of circulating miR-92a are found in the sera of lung cancer patients, and EVs isolated from these patients have a similar ability to increase HSCs activation and ECM protein expression. Our study reveals the sequential steps of liver PMN formation in lung cancer, providing critical mediators that prepare PMN in the liver, and identifies new targets that offer valuable options for diagnosis and therapeutic intervention.


Bone Marrow/metabolism , Extracellular Vesicles/genetics , Hepatic Stellate Cells/metabolism , Liver Neoplasms/secondary , Lung Neoplasms/pathology , MicroRNAs/genetics , Tumor Microenvironment , Animals , Apoptosis , Case-Control Studies , Cell Line, Tumor , Disease Models, Animal , Extracellular Vesicles/metabolism , Healthy Volunteers , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude , MicroRNAs/blood , Smad7 Protein/metabolism , Xenograft Model Antitumor Assays
6.
Breast Cancer Res ; 21(1): 23, 2019 02 12.
Article En | MEDLINE | ID: mdl-30755260

BACKGROUND: Metastasis is the major cause of death from breast cancer. Colonization and adaption of metastatic cells in distant organs is a rate-limiting step of the cancer spreading. The underlying mechanisms responsible for the colonization of breast cancer to lung metastatic niches are not fully understood. METHODS: Specific gene contributions to lung metastasis were identified by comparing gene profiles of 4T1 tumors metastasizing to various organs via microarray. The oncogenic properties CXCL17 were examined by in vivo spontaneous metastasis mouse model. The chemotactic activity of CXCL17 on CD11b+Gr-1+ myeloid-derived suppressor cells (MDSCs) was examined by both in vitro and in vivo models. The therapeutic effects of MDSC depletion and platelet-derived growth factor-BB (PDGF-BB) inhibition were examined by orthotic models. RESULTS: Here, we demonstrate that breast cancer cells secrete CXCL17, which increases the accumulation of CD11b+Gr-1+ MDSCs in the lungs. Metastatic lung-infiltrating CD11b+Gr-1+ MDSCs induce angiogenesis in the lungs and facilitate cancer extravasation and survival that ultimately promote lung metastases. CXCL17 increases CD11b+Gr-1+ MDSCs to express PDGF-BB, which not only contributes to CD11b+Gr-1+ MDSC-mediated angiogenesis in the lung metastatic niche, but is also involved in the colonization of breast cancer. Consequently, both CD11b+Gr-1+ MDSC depletion and PDGF receptor inhibitor effectively prevents CXCL17-driven lung metastasis in breast cancer. More importantly, patients with high levels of CXCL17 have shorter distant metastasis-free and overall survival rates, indicators of poor prognosis. CONCLUSION: Our study reveals that MDSCs derived by CXCL17 contribute to the establishment of a lung metastatic niche by PDGF-BB secretion and provide a rationale for development of CXCL17 or PDGF-BB antagonists to inhibit or prevent lung metastasis in cases of breast cancer.


Becaplermin/metabolism , Breast Neoplasms/pathology , Chemokines/metabolism , Lung Neoplasms/pathology , Myeloid-Derived Suppressor Cells/pathology , Animals , Breast Neoplasms/blood , Breast Neoplasms/immunology , Breast Neoplasms/mortality , CD11b Antigen/metabolism , Cell Line, Tumor , Chemokines/blood , Chemokines, CXC/metabolism , Chemotaxis , Datasets as Topic , Female , Humans , Kaplan-Meier Estimate , Lung/pathology , Lung Neoplasms/secondary , Mice , Mice, Nude , Myeloid-Derived Suppressor Cells/immunology , Prognosis , Receptors, Chemokine/metabolism , Xenograft Model Antitumor Assays
7.
Mol Ther ; 26(2): 568-581, 2018 02 07.
Article En | MEDLINE | ID: mdl-29292163

Hypoxia, the most commonly observed characteristic in cancers, is implicated in the establishment of an immunosuppressive niche. Recent studies have indicated that extracellular vesicle (EV)-mediated cancer-stroma interactions are considered to play a critical role in the regulation of various cellular biological functions, with phenotypic consequences in recipient cells. However, the mechanisms underlying the relationship between EVs and hypoxia during cancer progression remain largely unknown. In this study, we found that EVs derived from hypoxic lung cancers increased M2-type polarization by miR-103a transfer. Decreased PTEN levels caused by hypoxic cancer-cell-derived EV miR-103a increased activation of AKT and STAT3 as well as expression of several immunosuppressive and pro-angiogeneic factors. In contrast, inhibition of miR-103a by an miRNA inhibitor effectively decreased hypoxic cancer-mediated M2-type polarization, improving the cytokine prolife of tumor infiltration macrophages. Macrophages received cancer-cell-derived EV miR-103a feedback to further enhance cancer progression and tumor angiogenesis. Finally, circulating EV miR-103a levels were higher in patients with lung cancer and closely associated with the M2 polarization. In conclusion, our results delineate a novel mechanism by which lung cancer cells induce immunosuppressive and pro-tumoral macrophages through EVs and inspire further research into the clinical application of EV inhibition or PTEN restoration for immunotherapy.


Hypoxia/genetics , Hypoxia/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Macrophages/metabolism , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , RNA Interference , 3' Untranslated Regions , Cell Line, Tumor , Cytokines/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Macrophage Activation/genetics , Macrophage Activation/immunology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism
8.
J Biomed Sci ; 21: 46, 2014 May 16.
Article En | MEDLINE | ID: mdl-24885237

BACKGROUND: Intermittent hypoxia (IH) plays a critical role in sleep breathing disorder-associated hippocampus impairments, including neurocognitive deficits, irreversible memory and learning impairments. IH-induced neuronal injury in the hippocampus may result from reduced precursor cell proliferation and the relative numbers of postmitotic differentiated neurons. However, the mechanisms underlying IH-induced reactive oxygen species (ROS) generation effects on cell proliferation and neuronal differentiation remain largely unknown. RESULTS: ROS generation significantly increased after 1-4 days of IH without increased pheochromocytoma-12 (PC12) cell death, which resulted in increased protein phosphatase 2A (PP2A) mRNA and protein levels. After 3-4 days of IH, extracellular signal-regulated kinases 1/2 (ERK1/2) protein phosphorylation decreased, which could be reversed by superoxide dismutase (SOD), 1,10-phenanthroline (Phe), the PP2A phosphorylation inhibitors, okadaic acid (OKA) and cantharidin, and the ERK phosphorylation activator nicotine (p < 0.05). In particular, the significantly reduced cell proliferation and increased proportions of cells in the G0/G1 phase after 1-4 days of IH (p < 0.05), which resulted in decreased numbers of PC12 cells, could be reversed by treatment with SOD, Phe, PP2A inhibitors and an ERK activator. In addition, the numbers of nerve growth factor (NGF)-induced PC12 cells with neurite outgrowths after 3-4 days of IH were less than those after 4 days of RA, which was also reversed by SOD, Phe, PP2A inhibitors and an ERK activator. CONCLUSIONS: Our results suggest that IH-induced ROS generation increases PP2A activation and subsequently downregulates ERK1/2 activation, which results in inhibition of PC12 cell proliferation through G0/G1 phase arrest and NGF-induced neuronal differentiation.


Cell Differentiation/genetics , Enzyme Activation/genetics , Protein Phosphatase 2/biosynthesis , Sleep Wake Disorders/enzymology , Animals , Cell Proliferation/drug effects , Hippocampus/enzymology , Hypoxia/complications , Hypoxia/enzymology , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Growth Factor/pharmacology , PC12 Cells , Phosphorylation , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/genetics , Rats , Reactive Oxygen Species/metabolism , Sleep Wake Disorders/complications , Sleep Wake Disorders/pathology , Superoxide Dismutase/metabolism
...