Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Cell Genom ; : 100565, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38781966

ABSTRACT

Spatially resolved transcriptomics (SRT) technologies have revolutionized the study of tissue organization. We introduce a graph convolutional network with an attention and positive emphasis mechanism, termed BINARY, relying exclusively on binarized SRT data to accurately delineate spatial domains. BINARY outperforms existing methods across various SRT data types while using significantly less input information. Our study suggests that precise gene expression quantification may not always be essential, inspiring further exploration of the broader applications of spatially resolved binarized gene expression data.

2.
J Bone Oncol ; 45: 100598, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38585688

ABSTRACT

Previous studies illustrated that macrophage, a type of innate immune cell, plays critical roles in tumour progression and metastasis. Bone is the most frequent site of metastasis for several cancer types including breast, prostate, and lung. In bone metastasis, osteoclast, a macrophage subset specialized in bone resorption, was heavily investigated in the past. Recent studies illustrated that other macrophage subsets, e.g. monocyte-derived macrophages, and bone resident macrophages, promoted bone metastasis independent of osteoclast function. These novel mechanisms further improved our understanding of macrophage heterogeneity in the context of bone metastasis and illustrated new opportunities for future studies.

3.
Immunity ; 56(6): 1153-1154, 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37315527
4.
Life Sci Alliance ; 6(9)2023 09.
Article in English | MEDLINE | ID: mdl-37385752

ABSTRACT

The Hippo signalling pathway is a master regulator of cell growth, proliferation, and cancer. The transcriptional coregulators of the Hippo pathway, YAP and TAZ, are central in various cancers. However, how YAP and TAZ get activated in most types of cancers is not well understood. Here, we show that androgens activate YAP/TAZ via the androgen receptor (AR) in prostate cancer (PCa), and that this activation is differential. AR regulates YAP translation while inducing transcription of the TAZ encoding gene, WWTR1 Furthermore, we show that AR-mediated YAP/TAZ activation is regulated by the RhoA GTPases transcriptional mediator, serum response factor (SRF). Importantly, in prostate cancer patients, SRF expression positively correlates with TAZ and the YAP/TAZ target genes CYR61 and CTGF We demonstrate that YAP/TAZ are not essential for sustaining AR activity, however, targeting YAP/TAZ or SRF sensitize PCa cells to AR inhibition in anchorage-independent growth conditions. Our findings dissect the cellular roles of YAP, TAZ, and SRF in prostate cancer cells. Our data emphasize the interplay between these transcriptional regulators and their roles in prostate tumorigenesis and highlight how these insights might be exploited therapeutically.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Humans , Male , Androgens , Carcinogenesis , Prostate , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism
5.
Nat Nanotechnol ; 18(10): 1195-1204, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37349506

ABSTRACT

Optical imaging in the second near-infrared window (NIR-II, 1,000-1,700 nm) holds great promise for non-invasive in vivo detection. However, real-time dynamic multiplexed imaging remains challenging due to the lack of available fluorescence probes and multiplexing techniques in the ideal NIR-IIb (1,500-1,700 nm) 'deep-tissue-transparent' sub-window. Here we report on thulium-based cubic-phase downshifting nanoparticles (α-TmNPs) with 1,632 nm fluorescence amplification. This strategy was also validated for the fluorescence enhancement of nanoparticles doped with NIR-II Er3+ (α-ErNPs) or Ho3+ (α-HoNPs). In parallel, we developed a simultaneous dual-channel imaging system with high spatiotemporal synchronization and accuracy. The NIR-IIb α-TmNPs and α-ErNPs facilitated the non-invasive real-time dynamic multiplexed imaging of cerebrovascular vasomotion activity and the single-cell-level neutrophil behaviour in mouse subcutaneous tissue and ischaemic stroke model.


Subject(s)
Brain Ischemia , Nanoparticles , Stroke , Animals , Mice , Nanoparticles/chemistry , Fluorescent Dyes/chemistry , Optical Imaging
6.
J Exp Med ; 220(4)2023 04 03.
Article in English | MEDLINE | ID: mdl-36749798

ABSTRACT

Metastatic castration-resistant prostate cancer (PC) is the final stage of PC that acquires resistance to androgen deprivation therapies (ADT). Despite progresses in understanding of disease mechanisms, the specific contribution of the metastatic microenvironment to ADT resistance remains largely unknown. The current study identified that the macrophage is the major microenvironmental component of bone-metastatic PC in patients. Using a novel in vivo model, we demonstrated that macrophages were critical for enzalutamide resistance through induction of a wound-healing-like response of ECM-receptor gene expression. Mechanistically, macrophages drove resistance through cytokine activin A that induced fibronectin (FN1)-integrin alpha 5 (ITGA5)-tyrosine kinase Src (SRC) signaling cascade in PC cells. This novel mechanism was strongly supported by bioinformatics analysis of patient transcriptomics datasets. Furthermore, macrophage depletion or SRC inhibition using a novel specific inhibitor significantly inhibited resistant growth. Together, our findings elucidated a novel mechanism of macrophage-induced anti-androgen resistance of metastatic PC and a promising therapeutic approach to treat this deadly disease.


Subject(s)
Bone Neoplasms , Prostatic Neoplasms, Castration-Resistant , Male , Humans , Androgen Antagonists/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Cell Line, Tumor , Macrophages/metabolism , Receptors, Androgen/genetics , Nitriles/therapeutic use , Tumor Microenvironment
7.
IEEE Rev Biomed Eng ; 16: 499-513, 2023.
Article in English | MEDLINE | ID: mdl-35302938

ABSTRACT

Technologies for quantifying circulating tumour DNA (ctDNA) in liquid biopsies could enable real-time measurements of cancer progression, profoundly impacting patient care. Sequencing methods can be too complex and time-consuming for regular point-of-care monitoring, but nanotechnology offers an alternative, harnessing the unique properties of objects tens to hundreds of nanometres in size. This systematic review was performed to identify all examples of nanotechnology-based ctDNA detection and assess their potential for clinical use. Google Scholar, PubMed, Web of Science, Google Patents, Espacenet and Embase/MEDLINE were searched up to 23rd March 2021. The review identified nanotechnology-based methods for ctDNA detection for which quantitative measures (e.g., limit of detection, LOD) were reported and biologically relevant samples were used. The pre-defined inclusion criteria were met by 66 records. LODs ranged from 10 zM to 50nM. 25 records presented an LOD of 10fM or below. Nanotechnology-based approaches could provide the basis for the next wave of advances in ctDNA diagnostics, enabling analysis at the point-of-care, but none are currently used clinically. Further work is needed in development and validation; trade-offs are expected between different performance measures e.g., number of sequences detected and time to result.


Subject(s)
Circulating Tumor DNA , Neoplasms , Humans , Circulating Tumor DNA/genetics , Biomarkers, Tumor/genetics , Nanotechnology , Liquid Biopsy/methods
8.
Cancers (Basel) ; 14(17)2022 Sep 05.
Article in English | MEDLINE | ID: mdl-36077870

ABSTRACT

Metastasis is the systemic manifestation of cancer and the main cause of death from breast cancer. In mouse models of lung metastases, recruitment of classical monocytes from blood to the lung and their differentiation to metastasis-associated macrophages (MAMs) facilitate cancer cell extravasation, survival and growth. Ablation of MAMs or their monocytic progenitors inhibits metastasis. We hypothesized that factors controlling macrophage polarization modulate tumor cell extravasation in the lung. We evaluated whether signaling by Th1 or Th2 cytokines in macrophages affected transendothelial migration of tumor cells in vitro. Interferon gamma and LPS inhibited macrophage-dependent tumor cell extravasation while the Th2 cytokine interleukin-4 (IL4) enhanced this process. We demonstrated that IL4 receptor (IL4rα)-null mice developed fewer and smaller lung metastasis in E0771-LG mammary cancer models of this disease. Adoptive transfer of wild-type monocytes to IL4rα-deficient mice partially rescued this phenotype. IL4 signaling in macrophages controlled the expression of the chemokine receptor CXCR2, necessary for IL4-mediated tumor cell extravasation in vitro. Furthermore, IL4 signaling in macrophages regulated the transcript abundance of several other genes already causally associated with mammary cancer lung metastasis including Ccl2, Csf1, Ccr1, Hgf and Flt1. The central role of IL4 signaling in MAMs was confirmed by high-resolution intravital imaging of the lung in mice at the time of metastatic seeding, which showed reduced physical interaction between tumor cells and IL4rα-deficient macrophages. This interaction with wild-type MAMs enhanced tumor cell survival and seeding, which was lost in the IL4rα mice. These data indicate that IL4 signaling in monocytes and macrophages is key during seeding and growth of breast metastasis in the lung, as it regulates pro-tumoral paracrine signaling between cancer cells and macrophages.

9.
Nat Commun ; 13(1): 4674, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35945217

ABSTRACT

The MYC oncogene is a potent driver of growth and proliferation but also sensitises cells to apoptosis, which limits its oncogenic potential. MYC induces several biosynthetic programmes and primary cells overexpressing MYC are highly sensitive to glutamine withdrawal suggesting that MYC-induced sensitisation to apoptosis may be due to imbalance of metabolic/energetic supply and demand. Here we show that MYC elevates global transcription and translation, even in the absence of glutamine, revealing metabolic demand without corresponding supply. Glutamine withdrawal from MRC-5 fibroblasts depletes key tricarboxylic acid (TCA) cycle metabolites and, in combination with MYC activation, leads to AMP accumulation and nucleotide catabolism indicative of energetic stress. Further analyses reveal that glutamine supports viability through TCA cycle energetics rather than asparagine biosynthesis and that TCA cycle inhibition confers tumour suppression on MYC-driven lymphoma in vivo. In summary, glutamine supports the viability of MYC-overexpressing cells through an energetic rather than a biosynthetic mechanism.


Subject(s)
Apoptosis , Glutamine , Apoptosis/genetics , Cell Line, Tumor , Citric Acid Cycle , Fibroblasts/metabolism , Glutamine/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
10.
Trends Immunol ; 43(7): 546-563, 2022 07.
Article in English | MEDLINE | ID: mdl-35690521

ABSTRACT

Tumor-associated macrophages (TAMs) have multiple potent functions in cancer and, thus, represent important therapeutic targets. These diverse functions highlight the heterogenous nature of TAMs. Recent single cell omics technologies have significantly advanced our understanding of the molecular diversity of TAMs. However, a unifying nomenclature of TAM diversity and annotation of their molecular signatures is lacking. Here, we review recent major studies of single cell transcriptome, epigenome, metabolome, and spatial omics of cancer with a specific focus on TAMs. We also propose a consensus model of TAM diversity and present avenues for future research.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Macrophages , Neoplasms/therapy
11.
Small Methods ; 6(2): e2100966, 2022 02.
Article in English | MEDLINE | ID: mdl-35174992

ABSTRACT

With increasing interest in applying recombinant monoclonal antibodies (mAbs) in human medicine, engineered mAb fragments with reduced size and improved stability are in demand to overcome current limitations in clinical use. Herein, a novel Fab-like antibody fragment generated via an in silico-based engineering approach where the CH1 and CL domains of Fab are replaced by the IgG1 CH3 domains is described. This construct, designated as FabCH3, maintains the natural N-terminus and C-terminus of IgG antibody, can be expressed at a high level in bacterial cells and, importantly, exhibits much higher stability and affinity than the parental Fab when tested in a mesothelin-specific Fab m912, as well as a vascular endothelial growth factor A (VEGFA)-specific Fab Ranibizumab (in vivo). The high-resolution crystal structures of m912 FabCH3 and m912 Fab are determined, and the comparative analysis reveals more rigid structures in both constant domains and complementarity-determining regions of FabCH3, explaining its enhanced stability and affinity. Overall, the stabilized FabCH3 described in this report provides a versatile platform for engineering Fab-like antibody fragments with higher stability and antigen-binding affinity that can be used as a distinct class of antibody therapeutics.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Immunoglobulin Fab Fragments/administration & dosage , Immunoglobulin G/chemistry , Mesothelin/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antibody Affinity , Computer Simulation , Drug Design , Drug Stability , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/pharmacology , Male , Mesothelin/chemistry , Mice , Models, Molecular , Molecular Docking Simulation , Protein Conformation , Protein Domains , Ranibizumab/administration & dosage , Ranibizumab/chemistry , Ranibizumab/pharmacology
12.
Oxid Med Cell Longev ; 2021: 5806602, 2021.
Article in English | MEDLINE | ID: mdl-34721759

ABSTRACT

The bottleneck arising from castration-resistant prostate cancer (CRPC) treatment is its high metastasis potential and antiandrogen drug resistance, which severely affects survival time of prostate cancer (PCa) patients. Secreted phosphoprotein 1 (SPP1) is a cardinal mediator of tumor-associated inflammation and facilitates metastasis. In our previous study, we firstly revealed SPP1 was a potential hub signature for predicting metastatic CRPC (mCRPC) development. Herein, we integrated multiple databases to explore the association of SPP1 expression with prognosis, survival, and metastatic levels in CRPC progression and investigated SPP1 expression in PCa tissues and cell lines. Next, PCa cell lines with overexpression or depletion of SPP1 were established to study the effect of SPP1 on enzalutamide sensitivity and adhesion and migration of prostate cancer cell lines and further explore the underlying regulatory mechanisms. Bioinformatics analysis, polymerase chain reaction (PCR), immunohistochemical staining, and western blot results suggested SPP1 upregulation had strong relationship with the malignant progression of CRPC and enzalutamide resistance. SPP1 knockdown enhanced enzalutamide sensitivity and repressed invasion and migration of prostate cancer cells. Importantly, upregulating SPP1 promoted, while silencing SPP1 attenuated epithelial-mesenchymal-transition (EMT). Our results further demonstrated that SPP1 overexpression maintains the activation of PI3K/AKT and ERK1/2 signaling pathways. Overall, our findings unraveled the functional role and clinical significance of SPP1 in PCa progression and help to discover new potential targets against mCRPC.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Benzamides/pharmacology , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nitriles/pharmacology , Osteopontin/metabolism , Phenylthiohydantoin/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Invasiveness , Osteopontin/genetics , PC-3 Cells , Prostatic Neoplasms, Castration-Resistant/enzymology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Signal Transduction
13.
Cancer Res ; 81(21): 5438-5450, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34417202

ABSTRACT

Despite the approval of several multikinase inhibitors that target SRC and the overwhelming evidence of the role of SRC in the progression and resistance mechanisms of many solid malignancies, inhibition of its kinase activity has thus far failed to improve patient outcomes. Here we report the small molecule eCF506 locks SRC in its native inactive conformation, thereby inhibiting both enzymatic and scaffolding functions that prevent phosphorylation and complex formation with its partner FAK. This mechanism of action resulted in highly potent and selective pathway inhibition in culture and in vivo. Treatment with eCF506 resulted in increased antitumor efficacy and tolerability in syngeneic murine cancer models, demonstrating significant therapeutic advantages over existing SRC/ABL inhibitors. Therefore, this mode of inhibiting SRC could lead to improved treatment of SRC-associated disorders. SIGNIFICANCE: Small molecule-mediated inhibition of SRC impairing both catalytic and scaffolding functions confers increased anticancer properties and tolerability compared with other SRC/ABL inhibitors.


Subject(s)
Bone Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Focal Adhesion Kinase 1/antagonists & inhibitors , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Small Molecule Libraries/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Apoptosis , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Conformation , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , src-Family Kinases/chemistry , src-Family Kinases/metabolism
14.
Sci Adv ; 7(7)2021 02.
Article in English | MEDLINE | ID: mdl-33579710

ABSTRACT

The gut microbiota fundamentally regulates intestinal homeostasis and disease partially through mechanisms that involve modulation of regulatory T cells (Tregs), yet how the microbiota-Treg cross-talk is physiologically controlled is incompletely defined. Here, we report that prostaglandin E2 (PGE2), a well-known mediator of inflammation, inhibits mucosal Tregs in a manner depending on the gut microbiota. PGE2 through its receptor EP4 diminishes Treg-favorable commensal microbiota. Transfer of the gut microbiota that was modified by PGE2-EP4 signaling modulates mucosal Treg responses and exacerbates intestinal inflammation. Mechanistically, PGE2-modified microbiota regulates intestinal mononuclear phagocytes and type I interferon signaling. Depletion of mononuclear phagocytes or deficiency of type I interferon receptor diminishes PGE2-dependent Treg inhibition. Together, our findings provide emergent evidence that PGE2-mediated disruption of microbiota-Treg communication fosters intestinal inflammation.


Subject(s)
Gastrointestinal Microbiome , T-Lymphocytes, Regulatory , Dinoprostone/pharmacology , Humans , Inflammation , Receptors, Prostaglandin E, EP2 Subtype
15.
J Exp Med ; 217(11)2020 11 02.
Article in English | MEDLINE | ID: mdl-32780802

ABSTRACT

Bone metastasis is the major cause of death in breast cancer. The lack of effective treatment suggests that disease mechanisms are still largely unknown. As a key component of the tumor microenvironment, macrophages promote tumor progression and metastasis. In this study, we found that macrophages are abundant in human and mouse breast cancer bone metastases. Macrophage ablation significantly inhibited bone metastasis growth. Lineage tracking experiments indicated that these macrophages largely derive from Ly6C+CCR2+ inflammatory monocytes. Ablation of the chemokine receptor, CCR2, significantly inhibited bone metastasis outgrowth and prolonged survival. Immunophenotyping identified that bone metastasis-associated macrophages express high levels of CD204 and IL4R. Furthermore, monocyte/macrophage-restricted IL4R ablation significantly inhibited bone metastasis growth, and IL4R null mutant monocytes failed to promote bone metastasis outgrowth. Together, this study identified a subset of monocyte-derived macrophages that promote breast cancer bone metastasis in an IL4R-dependent manner. This suggests that IL4R and macrophage inhibition can have potential therapeutic benefit against breast cancer bone disease.


Subject(s)
Bone Neoplasms/immunology , Bone Neoplasms/secondary , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Macrophages/immunology , Adult , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/immunology , Cohort Studies , Disease Models, Animal , Female , Humans , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Middle Aged , Receptors, CCR2/genetics , Receptors, Cell Surface/genetics
16.
Acta Pharmacol Sin ; 40(9): 1228-1236, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31028291

ABSTRACT

Bufalin, the major active component of the traditional Chinese medicine ChanSu obtained from the skin and parotid venom glands of toads, has long been known as an anticancer agent. Recent studies show that microRNAs (miRs) are involved in the anticancer activities of bufalin, while long non-coding RNAs (lncRNAs) are known to interact with miRNAs to regulate various biological functions. In this paper, we investigated the possible network related to the antimetastatic effect of bufalin in prostate cancer (PCa) cells. We demonstrated that bufalin (0.05-10 µM) dose-dependently suppressed the proliferation of prostate cancer DU145 and PC3 cells with IC50 values of 0.89 and 1.28 µM, respectively. Furthermore, bufalin treatment significantly suppressed the cell migration and invasion. To explore the role of lncRNAs in the antimetastatic activity of bufalin, we used an lncRNA microarray and found that HOX transcript antisense RNA (HOTAIR) was the most markedly downregulated lncRNA in bufalin-treated PCa cells. Overexpression of HOTAIR counteracted the suppressing effects of bufalin on DU145 and PC3 cells. We then predicted and verified that HOTAIR upregulated FGFR1 expression by sponging miR-520b in PCa cells. In 40 patients with PCa bone metastasis, we used in situ hybridization or immunohistochemical assay to assess the HOTAIR and FGFR1 expression, which revealed that both HOTAIR and FGFR1 expression were significantly higher in bone metastasis tissues than in the primary PCa tissues. In addition, the level of serum HOTAIR was positively associated with the levels of serum bone metabolic markers (CTx, OST, B-ALP and PINP) and may serve as a reasonable biomarker for PCa bone metastasis. Taken together, this is the first study revealing that HOTAIR promotes PCa bone metastasis, and bufalin may be a promising candidate for the treatment of this disease.


Subject(s)
Antineoplastic Agents/pharmacology , Bufanolides/pharmacology , Cell Movement/drug effects , MicroRNAs/metabolism , Prostatic Neoplasms/metabolism , RNA, Long Noncoding/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Prostatic Neoplasms/drug therapy , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Up-Regulation/drug effects
17.
Pharmacol Res ; 144: 235-244, 2019 06.
Article in English | MEDLINE | ID: mdl-31028902

ABSTRACT

Metastasis to bone frequently occurs in majority of patients with advanced breast cancer and prostate cancer, leading to devastating skeletal-related events and substantially reducing the survival of patients. Currently, the crosstalk between tumor cells and the bone stromal compartment was widely investigated for bone metastasis and the resistance to many conventional therapeutic methods. Osteopontin (OPN), also known as SPP1 (secreted phosphoprotein 1), a secreted and chemokine-like glyco-phosphoprotein is involved in tumor progression such as cell proliferation, angiogenesis, and metastasis. The expression of OPN in tumor tissue and plasma has been clinically proved to be correlated to poor prognosis and shortened survival in patients with breast cancer and prostate cancer. This review summarizes the multifaceted roles that OPN plays in bone microenvironment and drug resistance, with emphasis on breast and prostate cancers, via binding to αvß3 integrin and CD44 receptor and inducing signaling cascades. We further discuss the promising therapeutic strategy for OPN targeting, mainly inhibiting OPN at transcriptional or protein level or blocking it binding to receptor or its downstream signaling pathways. The comprehending of the function of OPN in bone microenvironment is crucial for the development of novel biomarker and potential therapeutic target for the diagnosis and treatment of bone metastasis and against the emergence of drug resistance in advanced cancers.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Osteopontin/metabolism , Prostatic Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Female , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Tumor Microenvironment/drug effects
18.
Chem Sci ; 11(5): 1368-1374, 2019 Dec 18.
Article in English | MEDLINE | ID: mdl-34123261

ABSTRACT

Keratin 1 (KRT1) is overexpressed in squamous carcinomas and associated with aggressive pathologies in breast cancer. Herein we report the design and preparation of the first Trp-based red fluorogenic amino acid, which is synthetically accessible in a few steps and displays excellent photophysical properties, and its application in a minimally-disruptive labelling strategy to prepare a new fluorogenic cyclopeptide for imaging of KRT1+ cells in whole intact tumour tissues.

19.
Oncotarget ; 9(38): 25294-25303, 2018 May 18.
Article in English | MEDLINE | ID: mdl-29861872

ABSTRACT

BACKGROUND: Tumor associated macrophages (TAMs) have multifaceted roles in the development of many tumor types. However, the prognostic value of TAMs in bladder cancer is still not conclusive. EXPERIMENTAL DESIGN: This review evaluated the prognostic value of TAMs density in bladder cancer by reviewing published literatures and integrating the results via a meta-analysis. A systematic search was conducted in PubMed, Embase and Chinese National Knowledge Infrastructure (CNKI), WanFang, and Web of Science databases for relevant studies. Overall survival (OS), relapse free survival (RFS), disease specific survival (DSS), and progression free survival (PFS) were assessed in bladder cancer patients. RESULTS: The pooled hazard ratios (HRs) and 95% confidence intervals (CIs) indicated that TAMs identified with CD68 alone have no significant correlation with OS (HR = 1.01, 95% CI = 1.00-1.02), RFS (HR = 0.99, 95% CI = 0.91-1.06), or PFS (HR = 1.19, 95% CI = 0.70-1.68) in bladder cancer patients. Subgroup analyses involved with Bacillus Calmette Guerin (BCG) treatment or sample locations either showed that CD68+ TAMs presented no prognostic value with regard to OS in bladder cancer patients. However, TAMs detected by CD163 are significantly correlated with poor RFS in bladder cancer patients (HR = 1.54, 95% CI = 1.16-1.92). CONCLUSIONS: Our data indicated that TAMs identified only with CD68 have no significant correlation with the prognosis and clinicopathological parameters of bladder cancer patients. However, TAMs detected with CD163 could serve as a prognostic marker for bladder cancer patients. These findings invite further research on the role of TAM subsets in bladder cancer patients.

20.
Int J Mol Sci ; 19(4)2018 Apr 09.
Article in English | MEDLINE | ID: mdl-29642534

ABSTRACT

Bone metastasis is the most advanced stage of many cancers and indicates a poor prognosis for patients due to resistance to anti-tumor therapies. The establishment of metastasis within the bone is a multistep process. To ensure survival within the bone marrow, tumor cells must initially colonize a niche in which they can enter dormancy. Subsequently, reactivation permits the proliferation and growth of the tumor cells, giving rise to a macro-metastasis displayed clinically as a bone metastatic lesion. Here, we review the evidences that suggest mesenchymal stromal cells play an important role in each of these steps throughout the development of bone metastasis. Similarities between the molecular mechanisms implicated in these processes and those involved in the homeostasis of the bone indicate that the metastatic cells may exploit the homeostatic processes to their own advantage. Identifying the molecular interactions between the mesenchymal stromal cells and tumor cells that promote tumor development may offer insight into potential therapeutic targets that could be utilized to treat bone metastasis.


Subject(s)
Bone Neoplasms/pathology , Bone Neoplasms/secondary , Mesenchymal Stem Cells/physiology , Animals , Cell Proliferation , Homeostasis , Humans , Stem Cell Niche , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...