Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 995
Filter
1.
BMC Cancer ; 24(1): 1034, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169294

ABSTRACT

BACKGROUND: Melanoma is a highly aggressive form of skin cancer. The existence of cancer stem cells (CSCs) and tumor immune evasion are two major causes of melanoma progression, but no effective treatment has been found at present. Astragalus polysaccharide (APS) is a principal active component derived from Astragalus membranaceus, showing anti-tumor effects in various tumors including melanoma. However, the underlying mechanism is still unclear. METHODS: The regulation of APS on self-renewal ability and CSC markers expression in melanoma stem cells (MSCs) was measured by tumor sphere formation and tumorigenicity assays, RT-qPCR, and western blot. Flow cytometry was conducted to evaluate the activation of immune system by APS in melanoma mice. Further, the mechanism was explored based on PD-L1 overexpression and knock-down B16 cells. RESULTS: APS attenuated the tumor sphere formation of MSCs in vitro as well as the tumorigenicity in vivo. It also decreased the expression of CD133, BMI1 and CD47. Based on the PD-L1 overexpression and knock-down B16 cells, it was confirmed that APS inhibited the induction of MSCs by down-regulating PD-L1 expression. Meanwhile, APS increased the infiltration of CD4+ and CD8+T cells in tumor tissues because of its inhibitory effect on PD-L1. CONCLUSIONS: APS inhibited MSC induction and overcame tumor immune evasion through reducing PD-L1 expression. This study provided compelling evidence that APS could be a prospective therapeutic agent for treating melanoma.


Subject(s)
B7-H1 Antigen , Melanoma, Experimental , Neoplastic Stem Cells , Polysaccharides , B7-H1 Antigen/metabolism , Animals , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/immunology , Mice , Polysaccharides/pharmacology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Tumor Escape/drug effects , Cell Line, Tumor , Mice, Inbred C57BL , Humans , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Melanoma/immunology , Astragalus Plant/chemistry , Immune Evasion
2.
Sci Rep ; 14(1): 19560, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39174579

ABSTRACT

Three-dimensional (3D) bioprinting culture models capable of reproducing the pathological architecture of diseases are increasingly advancing. In this study, 3D scaffolds were created using extrusion-based bioprinting method with alginate, gelatin, and hyaluronic acid to investigate the effects of hyaluronic acid on the physical properties of the bioscaffold as well as on the formation of liver cancer spheroids. Conformational analysis, rheological characterization, and swelling-degradation tests were performed to characterize the scaffolds. After generating spheroids from hepatocellular carcinoma cells on the 3D scaffolds, cell viability and proliferation assays were performed. Flow cytometry and immunofluorescence microscopy were used into examine the expression of albumin, CD44, and E-cadherin to demonstrate functional capability and maturation levels of the spheroid-forming cells. The results show that hyaluronic acid in the scaffolds correlates with spheroid formation and provides high survival rates. It is also associated with an increase in CD44 expression and a decrease in E-cadherin, while there is no significant change in the albumin expression in the cells. Overall, the findings demonstrate that hyaluronic acid in a 3D hydrogel scaffold supports spheroid formation and may induce stemness. We present a promising 3D scaffold model for enhancing liver cancer spheroid formation and mimicking solid tumors. This model also has the potential for further studies to examine stem cell properties in 3D models.


Subject(s)
Hyaluronan Receptors , Hyaluronic Acid , Neoplastic Stem Cells , Spheroids, Cellular , Tissue Scaffolds , Hyaluronic Acid/pharmacology , Hyaluronic Acid/chemistry , Humans , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Tissue Scaffolds/chemistry , Hyaluronan Receptors/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Cell Survival/drug effects , Cadherins/metabolism , Cell Proliferation/drug effects , Bioprinting/methods , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Gelatin/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology
3.
Oncoimmunology ; 13(1): 2393442, 2024.
Article in English | MEDLINE | ID: mdl-39175947

ABSTRACT

The inflammatory tumor microenvironment (TME) is a key driver for tumor-promoting processes. Tumor-associated macrophages are one of the main immune cell types in the TME and their increased density is related to poor prognosis in prostate cancer. Here, we investigated the influence of pro-inflammatory (M1) and immunosuppressive (M2) macrophages on prostate cancer lineage plasticity. Our findings reveal that M1 macrophage secreted factors upregulate genes related to stemness while downregulating genes associated with androgen response in prostate cancer cells. The expression of cancer stem cell (CSC) plasticity markers NANOG, KLF4, SOX2, OCT4, and CD44 was stimulated by the secreted factors from M1 macrophages. Moreover, AR and its target gene PSA were observed to be suppressed in LNCaP cells treated with secreted factors from M1 macrophages. Inhibition of NFκB signaling using the IKK16 inhibitor resulted in downregulation of NANOG, SOX2, and CD44 and CSC plasticity. Our study highlights that the secreted factors from M1 macrophages drive prostate cancer cell plasticity by upregulating the expression of CSC plasticity markers through NFκB signaling pathway.


Subject(s)
Hyaluronan Receptors , Kruppel-Like Factor 4 , Macrophages , NF-kappa B , Nanog Homeobox Protein , Neoplastic Stem Cells , Prostatic Neoplasms , SOXB1 Transcription Factors , Signal Transduction , Male , Humans , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/genetics , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/genetics , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Kruppel-Like Factor 4/metabolism , NF-kappa B/metabolism , Cell Line, Tumor , Macrophages/metabolism , Up-Regulation , Tumor Microenvironment/immunology , Cell Plasticity/genetics , Gene Expression Regulation, Neoplastic , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Animals , Mice
4.
Mol Biol Rep ; 51(1): 912, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39153092

ABSTRACT

Glioblastoma is the most aggressive brain cancer with an unfavorable prognosis for patient survival. Glioma stem cells, a subpopulation of cancer cells, drive tumor initiation, self-renewal, and resistance to therapy and, together with the microenvironment, play a crucial role in glioblastoma maintenance and progression. Neurotransmitters such as noradrenaline, dopamine, and serotonin have contrasting effects on glioblastoma development, stimulating or inhibiting its progression depending on the cellular context and through their action on glioma stem cells, perhaps changing the epigenetic landscape. Recent studies have revealed that serotonin and dopamine induce chromatin modifications related to transcriptional plasticity in the mammalian brain and possibly in glioblastoma; however, this topic still needs to be explored because of its potential implications for glioblastoma treatment. Also, it is essential to consider that neurotransmitters' effects depend on the tumor's microenvironment since it can significantly influence the response and behavior of cancer cells. This review examines the possible role of neurotransmitters as regulators of glioblastoma development, focusing on their impact on the chromatin of glioma stem cells.


Subject(s)
Brain Neoplasms , Chromatin , Glioblastoma , Neoplastic Stem Cells , Neurotransmitter Agents , Tumor Microenvironment , Humans , Glioblastoma/metabolism , Glioblastoma/genetics , Glioblastoma/pathology , Neurotransmitter Agents/metabolism , Chromatin/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Microenvironment/genetics , Epigenesis, Genetic , Dopamine/metabolism , Animals , Serotonin/metabolism , Gene Expression Regulation, Neoplastic
5.
Cell Death Dis ; 15(8): 597, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39154024

ABSTRACT

The metastasis-associated protein (MTA) family plays a crucial role in the development of breast cancer, a common malignancy with a high incidence rate among women. However, the mechanism by which each member of the MTA family contributes to breast cancer progression is poorly understood. In this study, we aimed to investigate the roles of MTA1, MTA3, and tripartite motif-containing 21 (TRIM21) in the proliferation, invasion, epithelial-mesenchymal transition (EMT), and stem cell-like properties of breast cancer cells in vivo and in vitro. The molecular mechanisms of the feedback loop between MTA1 and MTA3/TRIM21 regulated by estrogen were explored using Chromatin immunoprecipitation (ChIP), luciferase reporter, immunoprecipitation (IP), and ubiquitination assays. These findings demonstrated that MTA1 acts as a driver to promote the progression of breast cancer by repressing the transcription of tumor suppressor genes, including TRIM21 and MTA3. Conversely, MTA3 inhibited MTA1 transcription and TRIM21 regulated MTA1 protein stability in breast cancer. Estrogen disrupted the balance between MTA1 and MTA3, as well as between MTA1 and TRIM21, thereby affecting stemness and the EMT processes in breast cancer. These findings suggest that MTA1 plays a vital role in stem cell fate and the hierarchical regulatory network of EMT through negative feedback loops with MTA3 or TRIM21 in response to estrogen, supporting MTA1, MTA3, and TRIM21 as potential prognostic biomarkers and MTA1 as a treatment target for future breast cancer therapies.


Subject(s)
Breast Neoplasms , Epithelial-Mesenchymal Transition , Estrogens , Histone Deacetylases , Neoplastic Stem Cells , Repressor Proteins , Trans-Activators , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Repressor Proteins/metabolism , Repressor Proteins/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Estrogens/pharmacology , Estrogens/metabolism , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Animals , Gene Expression Regulation, Neoplastic/drug effects , Mice , Cell Proliferation/drug effects , Cell Line, Tumor , Feedback, Physiological/drug effects , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Mice, Nude , MCF-7 Cells , Mice, Inbred BALB C , Neoplasm Proteins
6.
Sci Adv ; 10(32): eadn1607, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39110807

ABSTRACT

Glioblastoma (GBM) is the most prevalent and aggressive malignant primary brain tumor. GBM proximal to the lateral ventricles (LVs) is more aggressive, potentially because of subventricular zone contact. Despite this, cross-talk between GBM and neural stem/progenitor cells (NSC/NPCs) is not well understood. Using cell-specific proteomics, we show that LV-proximal GBM prevents neuronal maturation of NSCs through induction of senescence. In addition, GBM brain tumor-initiating cells (BTICs) increase expression of cathepsin B (CTSB) upon interaction with NPCs. Lentiviral knockdown and recombinant protein experiments reveal that both cell-intrinsic and soluble CTSB promote malignancy-associated phenotypes in BTICs. Soluble CTSB stalls neuronal maturation in NPCs while promoting senescence, providing a link between LV-tumor proximity and neurogenesis disruption. Last, we show LV-proximal CTSB up-regulation in patients, showing the relevance of this cross-talk in human GBM biology. These results demonstrate the value of proteomic analysis in tumor microenvironment research and provide direction for new therapeutic strategies in GBM.


Subject(s)
Brain Neoplasms , Cathepsin B , Glioblastoma , Lateral Ventricles , Neural Stem Cells , Proteomics , Signal Transduction , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Cathepsin B/metabolism , Cathepsin B/genetics , Humans , Proteomics/methods , Lateral Ventricles/metabolism , Lateral Ventricles/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Animals , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Cell Line, Tumor , Neurogenesis , Mice , Tumor Microenvironment
7.
Genome Med ; 16(1): 98, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138551

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAFs) are the prominent cell type in the tumor microenvironment (TME), and CAF subsets have been identified in various tumors. However, how CAFs spatially coordinate other cell populations within the liver TME to promote cancer progression remains unclear. METHODS: We combined multi-region proteomics (6 patients, 24 samples), 10X Genomics Visium spatial transcriptomics (11 patients, 25 samples), and multiplexed imaging (92 patients, 264 samples) technologies to decipher the expression heterogeneity, functional diversity, spatial distribution, colocalization, and interaction of fibroblasts. The newly identified CAF subpopulation was validated by cells isolated from 5 liver cancer patients and in vitro functional assays. RESULTS: We identified a liver CAF subpopulation, marked by the expression of COL1A2, COL4A1, COL4A2, CTGF, and FSTL1, and named F5-CAF. F5-CAF is preferentially located within and around tumor nests and colocalizes with cancer cells with higher stemness in hepatocellular carcinoma (HCC). Multiplexed staining of 92 patients and the bulk transcriptome of 371 patients demonstrated that the abundance of F5-CAFs in HCC was associated with a worse prognosis. Further in vitro experiments showed that F5-CAFs isolated from liver cancer patients can promote the proliferation and stemness of HCC cells. CONCLUSIONS: We identified a CAF subpopulation F5-CAF in liver cancer, which is associated with cancer stemness and unfavorable prognosis. Our results provide potential mechanisms by which the CAF subset in the TME promotes the development of liver cancer by supporting the survival of cancer stem cells.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Hepatocellular , Liver Neoplasms , Neoplastic Stem Cells , Tumor Microenvironment , Humans , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Microenvironment/genetics , Proteomics/methods , Transcriptome , Gene Expression Regulation, Neoplastic , Genomics/methods , Cell Proliferation , Gene Expression Profiling , Cell Line, Tumor , Prognosis , Multiomics
8.
J Hematol Oncol ; 17(1): 67, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143574

ABSTRACT

BACKGROUND: The interleukin-1 receptor accessory protein (IL1RAP) is highly expressed on acute myeloid leukemia (AML) bulk blasts and leukemic stem cells (LSCs), but not on normal hematopoietic stem cells (HSCs), providing an opportunity to target and eliminate the disease, while sparing normal hematopoiesis. Herein, we report the activity of BIF002, a novel anti-IL1RAP/CD3 T cell engager (TCE) in AML. METHODS: Antibodies to IL1RAP were isolated from CD138+ B cells collected from the immunized mice by optoelectric positioning and single cell sequencing. Individual mouse monoclonal antibodies (mAbs) were produced and characterized, from which we generated BIF002, an anti-human IL1RAP/CD3 TCE using Fab arm exchange. Mutations in human IgG1 Fc were introduced to reduce FcγR binding. The antileukemic activity of BIF002 was characterized in vitro and in vivo using multiple cell lines and patient derived AML samples. RESULTS: IL1RAP was found to be highly expressed on most human AML cell lines and primary blasts, including CD34+ LSC-enriched subpopulation from patients with both de novo and relapsed/refractory (R/R) leukemia, but not on normal HSCs. In co-culture of T cells from healthy donors and IL1RAPhigh AML cell lines and primary blasts, BIF002 induced dose- and effector-to-target (E:T) ratio-dependent T cell activation and leukemic cell lysis at subnanomolar concentrations. BIF002 administered intravenously along with human T cells led to depletion of leukemic cells, and significantly prolonged survival of IL1RAPhigh MOLM13 or AML patient-derived xenografts with no off-target side effects, compared to controls. Of note, BiF002 effectively redirects T cells to eliminate LSCs, as evidenced by the absence of disease initiation in secondary recipients of bone marrow (BM) from BIF002+T cells-treated donors (median survival not reached; all survived > 200 days) compared with recipients of BM from vehicle- (median survival: 26 days; p = 0.0004) or isotype control antibody+T cells-treated donors (26 days; p = 0.0002). CONCLUSIONS: The novel anti-IL1RAP/CD3 TCE, BIF002, eradicates LSCs and significantly prolongs survival of AML xenografts, representing a promising, novel treatment for AML.


Subject(s)
Interleukin-1 Receptor Accessory Protein , Leukemia, Myeloid, Acute , Neoplastic Stem Cells , T-Lymphocytes , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/drug therapy , Humans , Animals , Mice , Interleukin-1 Receptor Accessory Protein/immunology , T-Lymphocytes/immunology , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/drug effects , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Mice, Inbred NOD
9.
Cell Death Dis ; 15(8): 554, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090090

ABSTRACT

Glioma is the most common primary brain tumor, and targeting glioma stem cells (GSCs) has become a key aspect of glioma treatment. In this study, we discovered a molecular network in which circRNA forms an R-loop structure with its parental gene to regulate the biological behavior of GSCs. Genes with abnormal expression in GSCs were screened using RNA-seq and circRNA microarray analyses. The study results showed that high expression of YTHDC1 in GSCs promoted the transportation of N6-methyladenosine (m6A)-modified circPOLR2B from the nucleus to the cytoplasm. Decreased circPOLR2B levels in the nucleus resulted in fewer R-loop structures formed with its parental gene POLR2B. This reduction in R-loop structures relieved the inhibitory effect on POLR2B transcription and upregulated PBX1 expression through alternative polyadenylation (APA) action, thereby promoting the malignant biological behavior of GSCs. Knockdown of YTHDC1, POLR2B, and PBX1 reduced xenograft tumor volume and prolonged the survival of nude mice. The YTHDC1/circPOLR2B/POLR2B/PBX1 axis plays a regulatory role in the biological behavior of GSCs, offering potential targets and novel strategies for the treatment of glioma.


Subject(s)
Glioma , Neoplastic Stem Cells , RNA, Circular , Animals , Humans , Mice , Adenosine/analogs & derivatives , Adenosine/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Cell Line, Tumor , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Methylation , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , RNA, Circular/genetics , RNA, Circular/metabolism
11.
J Exp Med ; 221(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39167075

ABSTRACT

Changes in mechanosensitive ion channels following radiation have seldom been linked to therapeutic sensitivity or specific factors involved in antitumor immunity. Here, in this study, we found that the mechanical force sensor, Piezo2, was significantly upregulated in tumor cells after radiation, and Piezo2 knockout in tumor cells enhanced tumor growth suppression by radiotherapy. Specifically, loss of Piezo2 in tumor cells induced their IL-15 expression via unleashing JAK2/STAT1/IRF-1 axis after radiation. This increase in IL-15 activates IL-15Rα on tumor-infiltrating CD8+ T cells, thereby leading to their augmented effector and stem cell-like properties, along with reduced terminal exhausted feature. Importantly, Piezo2 expression was negatively correlated with CD8 infiltration, as well as with radiosensitivity of patients with rectum adenocarcinoma receiving radiotherapy treatment. Together, our findings reveal that tumor cell-intrinsic Piezo2 induces radioresistance by dampening the IRF-1/IL-15 axis, thus leading to impaired CD8+ T cell-dependent antitumor responses, providing insights into the further development of combination strategies to treat radioresistant cancers.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-15 , Ion Channels , Radiation Tolerance , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Animals , Humans , Ion Channels/metabolism , Ion Channels/genetics , Radiation Tolerance/genetics , Mice , Interleukin-15/metabolism , Interleukin-15/genetics , Cell Line, Tumor , Janus Kinase 2/metabolism , Janus Kinase 2/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/genetics , Mice, Inbred C57BL , Female , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Male , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Signal Transduction
12.
Article in English | MEDLINE | ID: mdl-39091260

ABSTRACT

The recurrent nature of glioblastoma negatively impacts conventional treatment strategies leading to a growing need for nanomedicine. Nanotherapeutics, an approach designed to deliver drugs to specific sites, is experiencing rapid growth and gaining immense popularity. Having potential in reaching the hard-to-reach disease sites, this field has the potential to show high efficacy in combatting glioblastoma progression. The presence of glioblastoma stem cells (GSCs) is a major factor behind the poor prognosis of glioblastoma multiforme (GBM). Stemness potential, heterogeneity, and self-renewal capacity, are some of the properties that make GSCs invade across the distant regions of the brain. Despite advances in medical technology and MRI-guided maximal surgical resection, not all GSCs residing in the brain can be removed, leading to recurrent disease. The aggressiveness of GBM is often correlated with immune suppression, where the T-cells are unable to infiltrate the cancer initiating GSCs. Standard of care therapies, including surgery and chemotherapy in combination with radiation therapy, have failed to tackle all the challenges of the GSCs, making it increasingly important for researchers to develop strategies to tackle their growth and proliferation and reduce the recurrence of GBM. Here, we will focus on the advancements in the field of nanomedicine that has the potential to show positive impact in managing glioblastoma tumor microenvironment. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.


Subject(s)
Brain Neoplasms , Glioblastoma , Nanomedicine , Neoplastic Stem Cells , Glioblastoma/therapy , Glioblastoma/diagnostic imaging , Glioblastoma/drug therapy , Humans , Brain Neoplasms/therapy , Brain Neoplasms/drug therapy , Animals , Neoplasm Recurrence, Local , Immunosuppression Therapy , Neoplasm Invasiveness , Mice
13.
Nat Commun ; 15(1): 6608, 2024 Aug 04.
Article in English | MEDLINE | ID: mdl-39098906

ABSTRACT

The antitumor performance of PROteolysis-TArgeting Chimeras (PROTACs) is limited by its insufficient tumor specificity and poor pharmacokinetics. These disadvantages are further compounded by tumor heterogeneity, especially the presence of cancer stem-like cells, which drive tumor growth and relapse. Herein, we design a region-confined PROTAC nanoplatform that integrates both reactive oxygen species (ROS)-activatable and hypoxia-responsive PROTAC prodrugs for the precise manipulation of bromodomain and extraterminal protein 4 expression and tumor eradication. These PROTAC nanoparticles selectively accumulate within and penetrate deep into tumors via response to matrix metalloproteinase-2. Photoactivity is then reactivated in response to the acidic intracellular milieu and the PROTAC is discharged due to the ROS generated via photodynamic therapy specifically within the normoxic microenvironment. Moreover, the latent hypoxia-responsive PROTAC prodrug is restored in hypoxic cancer stem-like cells overexpressing nitroreductase. Here, we show the ability of region-confined PROTAC nanoplatform to effectively degrade BRD4 in both normoxic and hypoxic environments, markedly hindering tumor progression in breast and head-neck tumor models.


Subject(s)
Cell Cycle Proteins , Nanoparticles , Proteolysis , Transcription Factors , Humans , Proteolysis/drug effects , Animals , Nanoparticles/chemistry , Cell Line, Tumor , Mice , Transcription Factors/metabolism , Female , Cell Cycle Proteins/metabolism , Reactive Oxygen Species/metabolism , Prodrugs/pharmacology , Prodrugs/chemistry , Photochemotherapy/methods , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Mice, Nude , Xenograft Model Antitumor Assays , Tumor Microenvironment/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Nuclear Proteins/metabolism , Matrix Metalloproteinase 2/metabolism , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Bromodomain Containing Proteins
14.
Stem Cell Res Ther ; 15(1): 256, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39135143

ABSTRACT

BACKGROUND: One of major challenges in breast tumor therapy is the existence of breast cancer stem cells (BCSCs). BCSCs are a small subpopulation of tumor cells that exhibit characteristics of stem cells. BCSCs are responsible for progression, recurrence, chemoresistance and metastasis of breast cancer. Ca2+ signalling plays an important role in diverse processes in cancer development. However, the role of Ca2+ signalling in BCSCs is still poorly understood. METHODS: A highly effective 3D soft fibrin gel system was used to enrich BCSC-like cells from ER+ breast cancer lines MCF7 and MDA-MB-415. We then investigated the role of two Ca2+-permeable ion channels Orai1 and Orai3 in the growth and stemness of BCSC-like cells in vitro, and tumorigenicity in female NOD/SCID mice in vivo. RESULTS: Orai1 RNA silencing and pharmacological inhibition reduced the growth of BCSC-like cells in tumor spheroids, decreased the expression levels of BCSC markers, and reduced the growth of tumor xenografts in NOD/SCID mice. Orai3 RNA silencing also had similar inhibitory effect on the growth and stemness of BCSC-like cells in vitro, and tumor xenograft growth in vivo. Mechanistically, Orai1 and SPCA2 mediate store-operated Ca2+ entry. Knockdown of Orai1 or SPCA2 inhibited glycolysis pathway, whereas knockdown of Orai3 or STIM1 had no effect on glycolysis. CONCLUSION: We found that Orai1 interacts with SPCA2 to mediate store-independent Ca2+ entry, subsequently promoting the growth and tumorigenicity of BCSC-like cells via glycolysis pathway. In contrast, Orai3 and STIM1 mediate store-operated Ca2+ entry, promoting the growth and tumorigenicity of BCSC-like cells via a glycolysis-independent pathway. Together, our study uncovered a well-orchestrated mechanism through which two Ca2+ entry pathways act through distinct signalling axes to finely control the growth and tumorigenicity of BCSCs.


Subject(s)
Breast Neoplasms , Calcium Channels , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells , ORAI1 Protein , ORAI1 Protein/metabolism , ORAI1 Protein/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Humans , Animals , Female , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Mice , Calcium Channels/metabolism , Calcium Channels/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Signal Transduction , Calcium Signaling , MCF-7 Cells
15.
Database (Oxford) ; 20242024 Aug 13.
Article in English | MEDLINE | ID: mdl-39137906

ABSTRACT

Cancer stemness plays an important role in cancer initiation and progression, and is the major cause of tumor invasion, metastasis, recurrence, and poor prognosis. Non-coding RNAs (ncRNAs) are a class of RNA transcripts that generally cannot encode proteins and have been demonstrated to play a critical role in regulating cancer stemness. Here, we developed the ncStem database to record manually curated and predicted ncRNAs associated with cancer stemness. In total, ncStem contains 645 experimentally verified entries, including 159 long non-coding RNAs (lncRNAs), 254 microRNAs (miRNAs), 39 circular RNAs (circRNAs), and 5 other ncRNAs. The detailed information of each entry includes the ncRNA name, ncRNA identifier, disease, reference, expression direction, tissue, species, and so on. In addition, ncStem also provides computationally predicted cancer stemness-associated ncRNAs for 33 TCGA cancers, which were prioritized using the random walk with restart (RWR) algorithm based on regulatory and co-expression networks. The total predicted cancer stemness-associated ncRNAs included 11 132 lncRNAs and 972 miRNAs. Moreover, ncStem provides tools for functional enrichment analysis, survival analysis, and cell location interrogation for cancer stemness-associated ncRNAs. In summary, ncStem provides a platform to retrieve cancer stemness-associated ncRNAs, which may facilitate research on cancer stemness and offer potential targets for cancer treatment. Database URL: http://www.nidmarker-db.cn/ncStem/index.html.


Subject(s)
Neoplasms , Neoplastic Stem Cells , RNA, Untranslated , Humans , Neoplasms/genetics , Neoplasms/metabolism , RNA, Untranslated/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Databases, Nucleic Acid , Databases, Genetic , Data Curation/methods , MicroRNAs/genetics , MicroRNAs/metabolism
16.
J Vis Exp ; (209)2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39141548

ABSTRACT

Cellular quiescence is a state of growth arrest or slowed proliferation that is described in normal and cancer stem cells (CSCs). Quiescence may protect CSCs from antiproliferative chemotherapy drugs. In T-cell acute lymphoblastic leukemia (T-ALL) patient-derived xenograft (PDX) mouse models, quiescent cells are associated with treatment resistance and stemness. Cell proliferation dyes are popular tools for the tracking of cell division. The fluorescent dye is covalently anchored into amine groups on the membrane and macromolecules inside the cell. This allows for the tracking of labeled cells for up to 10 divisions, which can be resolved by flow cytometry. Ultimately, cells with the highest proliferation rates will have low dye retention, as it will be diluted with each cell division, while dormant, slower-dividing cells will have the highest retention. The use of cell proliferation dyes to isolate dormant cells has been optimized and described in T-ALL mouse models. Complementary to the existing mouse models, the rag2:Myc-derived zebrafish T-ALL model provides an excellent venue to interrogate self-renewal in T-ALL due to the high frequency of leukemic stem cells (LSCs) and the convenience of zebrafish for large-scale transplant experiments. Here, we describe the workflow for the staining of zebrafish T-ALL cells with a cell proliferation dye, optimizing the concentration of the dye for zebrafish cells, passaging successfully stained cells in vivo, and the collection of cells with varying levels of dye retention by live cell sorting from transplanted animals. Given the absence of well-established cell surface makers for LSCs in T-ALL, this approach provides a functional means to interrogate quiescent cells in vivo. For representative results, we describe the engraftment efficiency and the LSC frequency of high and low dye-retaining cells. This method can help investigate additional properties of quiescent cells, including drug response, transcriptional profiles, and morphology.


Subject(s)
Cell Proliferation , Fluorescent Dyes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Zebrafish , Animals , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Fluorescent Dyes/chemistry , Disease Models, Animal , Neoplastic Stem Cells/pathology , Flow Cytometry/methods
17.
Zhonghua Yu Fang Yi Xue Za Zhi ; 58(8): 1161-1170, 2024 Aug 06.
Article in Chinese | MEDLINE | ID: mdl-39142884

ABSTRACT

Central nervous system (CNS) tumors have complex causes and poor prognosis. Tumor heterogeneity is a major cause of treatment failure, and in-depth understanding of the biodiversity of CNS tumors is critical. Single-cell sequencing technology provides an opportunity to reveal the complex ecosystem of CNS tumors. In this study, we review the significance of single-cell sequencing in exploring the heterogeneity, diagnosis, treatment, and prognosis of CNS tumors from three aspects: tumor stem cells, tumor microenvironment, and cerebrospinal fluid. Although most of the findings have not been clinically applicable, they lay the foundation for the development of new guidelines for CNS tumors that help improve tumor prognosis and prevent tumor recurrence.


Subject(s)
Central Nervous System Neoplasms , Neoplastic Stem Cells , Single-Cell Analysis , Tumor Microenvironment , Humans , Central Nervous System Neoplasms/therapy , Central Nervous System Neoplasms/diagnosis , Single-Cell Analysis/methods , Prognosis
18.
Sci Rep ; 14(1): 18617, 2024 08 10.
Article in English | MEDLINE | ID: mdl-39127769

ABSTRACT

Endometrial cancer (EC), one of the most prevalent carcinomas in females, is associated with increasing mortality. We identified the CHD4 R975H mutation as a high-frequency occurrence in EC patients through a comprehensive survey of EC databases. Computational predictions suggest that this mutation profoundly impacts the structural and functional integrity of CHD4. Functional assays revealed that the CHD4 R975H mutation enhances EC cell invasion, proliferation, and colony formation, promoting a cancer stem cell (CSC)-like phenotype. RNA-seq analysis of cells expressing CHD4 R975H mutant revealed a transcriptomic landscape marked by the activation of several cancer-promoting signaling pathways, including TNF-α signaling via NF-κB, KRAS, P53, mTOR, TGF-ß, EGFR, Myc and growth factor signaling. Validation assays confirmed the activation of these pathways, further demonstrating that CHD4 R975H mutation induces stemness in EC cells and M2-like polarization of tumor-associated macrophages (TAMs). Our study elucidated the oncogenic role of CHD4 R975H mutation, highlighting its dual impact on facilitating cancer stemness and transforming TAMs into an immunosuppressive subtype. These findings contribute valuable insights into the molecular mechanisms driving EC progression and open avenues for targeted therapeutic interventions.


Subject(s)
Endometrial Neoplasms , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Neoplastic Stem Cells , Signal Transduction , Animals , Female , Humans , Mice , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Macrophages/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mutation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology
19.
J Transl Med ; 22(1): 737, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103915

ABSTRACT

BACKGROUND: Cancer stem-like cells (CSCs) play an important role in initiation and progression of aggressive cancers, including esophageal cancer. Natural killer (NK) cells are key effector lymphocytes of innate immunity that directly attack a wide variety of cancer cells. NK cell-based therapy may provide a new treatment option for targeting CSCs. In this study, we aimed to investigate the sensitivity of human esophageal CSCs to NK cell-mediated cytotoxicity. METHODS: CSCs were enriched from human esophageal squamous cell carcinoma cell lines via sphere formation culture. Human NK cells were selectively expanded from the peripheral blood of healthy donors. qRT-PCR, flow cytometry and ELISA assays were performed to examine RNA expression and protein levels, respectively. CFSE-labeled target cells were co-cultured with human activated NK cells to detect the cytotoxicity of NK cells by flow cytometry. RESULTS: We observed that esophageal CSCs were more resistant to NK cell-mediated cytotoxicity compared with adherent counterparts. Consistently, esophageal CSCs showed down-regulated expression of ULBP-1, a ligand for NK cells stimulatory receptor NKG2D. Knockdown of ULBP-1 resulted in significant inhibition of NK cell cytotoxicity against esophageal CSCs, whereas ULBP-1 overexpression led to the opposite effect. Finally, the pro-differentiation agent all-trans retinoic acid was found to enhance the sensitivity of esophageal CSCs to NK cell cytotoxicity. CONCLUSIONS: This study reveals that esophageal CSCs are more resistant to NK cells through down-regulation of ULBP-1 and provides a promising approach to promote the activity of NK cells targeting esophageal CSCs.


Subject(s)
Cytotoxicity, Immunologic , Down-Regulation , Esophageal Neoplasms , Killer Cells, Natural , Neoplastic Stem Cells , Humans , Killer Cells, Natural/immunology , Esophageal Neoplasms/pathology , Esophageal Neoplasms/immunology , Esophageal Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Down-Regulation/drug effects , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , GPI-Linked Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects
20.
Cell Metab ; 36(8): 1696-1710.e10, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39111285

ABSTRACT

Patients with high ALDH1A3-expressing glioblastoma (ALDH1A3hi GBM) show limited benefit from postoperative chemoradiotherapy. Understanding the mechanisms underlying such resistance in these patients is crucial for the development of new treatments. Here, we show that the interaction between ALDH1A3 and PKM2 enhances the latter's tetramerization and promotes lactate accumulation in glioblastoma stem cells (GSCs). By scanning the lactylated proteome in lactate-accumulating GSCs, we show that XRCC1 undergoes lactylation at lysine 247 (K247). Lactylated XRCC1 shows a stronger affinity for importin α, allowing for greater nuclear transposition of XRCC1 and enhanced DNA repair. Through high-throughput screening of a small-molecule library, we show that D34-919 potently disrupts the ALDH1A3-PKM2 interaction, preventing the ALDH1A3-mediated enhancement of PKM2 tetramerization. In vitro and in vivo treatment with D34-919 enhanced chemoradiotherapy-induced apoptosis of GBM cells. Together, our findings show that ALDH1A3-mediated PKM2 tetramerization is a potential therapeutic target to improve the response to chemoradiotherapy in ALDH1A3hi GBM.


Subject(s)
Glioblastoma , Thyroid Hormone-Binding Proteins , X-ray Repair Cross Complementing Protein 1 , Glioblastoma/metabolism , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Animals , Cell Line, Tumor , Mice , X-ray Repair Cross Complementing Protein 1/metabolism , X-ray Repair Cross Complementing Protein 1/genetics , Drug Resistance, Neoplasm/drug effects , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Membrane Proteins/metabolism , Carrier Proteins/metabolism , Thyroid Hormones/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Aldehyde Oxidoreductases , Oxidoreductases Acting on CH-NH Group Donors
SELECTION OF CITATIONS
SEARCH DETAIL