Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Int J Mol Sci ; 24(10)2023 May 12.
Article in English | MEDLINE | ID: mdl-37240029

ABSTRACT

Despite advances in treatment for metastatic melanoma patients, patients with liver metastasis have an unfavorable prognosis. A better understanding of the development of liver metastasis is needed. The multifunctional cytokine Transforming Growth Factor ß (TGF-ß) plays various roles in melanoma tumors and metastasis, affecting both tumor cells and cells from the surrounding tumor microenvironment. To study the role of TGF-ß in melanoma liver metastasis, we created a model to activate or repress the TGF-ß receptor pathway in vitro and in vivo in an inducible manner. For this, we engineered B16F10 melanoma cells to have inducible ectopic expression of a constitutively active (ca) or kinase-inactive (ki) TGF-ß receptor I, also termed activin receptor-like kinase (ALK5). In vitro, stimulation with TGF-ß signaling and ectopic caALK5 expression reduced B16F10 cell proliferation and migration. Contrasting results were found in vivo; sustained caALK5 expression in B16F10 cells in vivo increased the metastatic outgrowth in liver. Blocking microenvironmental TGF-ß did not affect metastatic liver outgrowth of both control and caALK5 expressing B16F10 cells. Upon characterizing the tumor microenvironment of control and caALk5 expressing B16F10 tumors, we observed reduced (cytotoxic) T cell presence and infiltration, as well as an increase in bone marrow-derived macrophages in caALK5 expressing B16F10 tumors. This suggests that caALK5 expression in B16F10 cells induces changes in the tumor microenvironment. A comparison of newly synthesized secreted proteins upon caALK5 expression by B16F10 cells revealed increased secretion of matrix remodeling proteins. Our results show that TGF-ß receptor activation in B16F10 melanoma cells can increase metastatic outgrowth in liver in vivo, possibly through remodeling of the tumor microenvironment leading to altered infiltration of immune cells. These results provide insights in the role of TGF-ß signaling in B16F10 liver metastasis and could have implications regarding the use of TGF-ß inhibitors for the treatment of melanoma patients with liver metastasis.


Subject(s)
Liver Neoplasms , Melanoma , Humans , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Cytokines , Transforming Growth Factor beta/metabolism , Cell Line, Tumor , Tumor Microenvironment
2.
J Am Soc Nephrol ; 31(5): 921-929, 2020 05.
Article in English | MEDLINE | ID: mdl-32354986

ABSTRACT

BACKGROUND: The utility of kidney organoids in regenerative medicine will rely on the functionality of the glomerular and tubular structures in these tissues. Recent studies have demonstrated the vascularization and subsequent maturation of human pluripotent stem cell-derived kidney organoids after renal subcapsular transplantation. This raises the question of whether the glomeruli also become functional upon transplantation. METHODS: We transplanted kidney organoids under the renal capsule of the left kidney in immunodeficient mice followed by the implantation of a titanium imaging window on top of the kidney organoid. To assess glomerular function in the transplanted human pluripotent stem cell-derived kidney tissue 1, 2, and 3 weeks after transplantation, we applied high-resolution intravital multiphoton imaging through the imaging window during intravenous infusion of fluorescently labeled low and high molecular mass dextran molecules or albumin. RESULTS: After vascularization, glomerular structures in the organoid displayed dextran and albumin size selectivity across their glomerular filtration barrier. We also observed evidence of proximal tubular dextran reuptake. CONCLUSIONS: Our results demonstrate that human pluripotent stem cell-derived glomeruli can develop an appropriate barrier function and discriminate between molecules of varying size. These characteristics together with tubular presence of low molecular mass dextran provide clear evidence of functional filtration. This approach to visualizing glomerular filtration function will be instrumental for translation of organoid technology for clinical applications as well as for disease modeling.


Subject(s)
Induced Pluripotent Stem Cells/transplantation , Kidney Glomerulus/metabolism , Organoids/transplantation , Albumins/metabolism , Animals , Dextrans/metabolism , Genes, Reporter , Humans , Induced Pluripotent Stem Cells/metabolism , Intravital Microscopy/methods , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Microscopy, Fluorescence, Multiphoton , Organoids/blood supply , Organoids/metabolism , Particle Size , Skin Window Technique , Time-Lapse Imaging/methods
3.
Exp Cell Res ; 381(1): 86-93, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30980788

ABSTRACT

Although two- and three-dimensional in vitro studies of breast tumor cell lines have increased our knowledge on tumor growth and metastasis formation, the complex in vivo microenvironment is not taken into consideration. The goal of our study was to illustrate the in vivo morphology and motility of widely used breast tumor cell lines. Intravital microscopy allows real-time visualization of individual cells inside tissues of living animals. We used this technique to study breast cancer migration in the complex orthotopic microenvironment. More specifically, we characterized cell morphology, cell-cell interactions, polarity and motility of mouse tumor cell lines 4T1 and mILC-1 and human tumor cell lines MDA-MB-231 and T47D. Almost all measured parameters were remarkably heterogeneous even between positions within the same tumor. Migrating tumor cells were circular in all tumor models, indicating predominantly amoeboid motility. This overview of the in vivo characteristics of mouse and human breast tumor cell lines illustrates their heterogeneity and complexity in real life, and additionally exemplifies caution should be taken to extrapolate in vitro assays of tumor invasiveness.


Subject(s)
Breast Neoplasms , Cell Line, Tumor , Animals , Breast Neoplasms/pathology , Cell Communication , Cell Movement , Cell Polarity , Female , Genetic Heterogeneity , Humans , Intravital Microscopy , Mice , Mice, Inbred BALB C , Neoplasm Transplantation
4.
Nature ; 507(7492): 362-365, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24531760

ABSTRACT

The rapid turnover of the mammalian intestinal epithelium is supported by stem cells located around the base of the crypt. In addition to the Lgr5 marker, intestinal stem cells have been associated with other markers that are expressed heterogeneously within the crypt base region. Previous quantitative clonal fate analyses have led to the proposal that homeostasis occurs as the consequence of neutral competition between dividing stem cells. However, the short-term behaviour of individual Lgr5(+) cells positioned at different locations within the crypt base compartment has not been resolved. Here we establish the short-term dynamics of intestinal stem cells using the novel approach of continuous intravital imaging of Lgr5- Confetti mice. We find that Lgr5(+) cells in the upper part of the niche (termed 'border cells') can be passively displaced into the transit-amplifying domain, after the division of proximate cells, implying that the determination of stem-cell fate can be uncoupled from division. Through quantitative analysis of individual clonal lineages, we show that stem cells at the crypt base, termed 'central cells', experience a survival advantage over border stem cells. However, through the transfer of stem cells between the border and central regions, all Lgr5(+) cells are endowed with long-term self-renewal potential. These findings establish a novel paradigm for stem-cell maintenance in which a dynamically heterogeneous cell population is able to function long term as a single stem-cell pool.


Subject(s)
Homeostasis , Intestinal Mucosa/cytology , Single-Cell Analysis , Stem Cells/cytology , Animals , Cell Division , Cell Lineage , Cell Survival , Clone Cells/cytology , Female , Male , Mice , Models, Biological , Molecular Imaging , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
5.
Int J Mol Sci ; 21(23)2020 Dec 03.
Article in English | MEDLINE | ID: mdl-33287463

ABSTRACT

Plaque angiogenesis and plaque hemorrhage are major players in the destabilization and rupture of atherosclerotic lesions. As these are dynamic processes, imaging of plaque angiogenesis, especially the integrity or leakiness of angiogenic vessels, can be an extremely useful tool in the studies on atherosclerosis pathophysiology. Visualizing plaque microvessels in 3D would enable us to study the architecture and permeability of adventitial and intimal plaque microvessels in advanced atherosclerotic lesions. We hypothesized that a comparison of the vascular permeability between healthy continuous and fenestrated as well as diseased leaky microvessels, would allow us to evaluate plaque microvessel leakiness. We developed and validated a two photon intravital microscopy (2P-IVM) method to assess the leakiness of plaque microvessels in murine atherosclerosis-prone ApoE3*Leiden vein grafts based on the quantification of fluorescent-dextrans extravasation in real-time. We describe a novel 2P-IVM set up to study vessels in the neck region of living mice. We show that microvessels in vein graft lesions are in their pathological state more permeable in comparison with healthy continuous and fenestrated microvessels. This 2P-IVM method is a promising approach to assess plaque angiogenesis and leakiness. Moreover, this method is an important advancement to validate therapeutic angiogenic interventions in preclinical atherosclerosis models.


Subject(s)
Intravital Microscopy , Microvessels/metabolism , Microvessels/pathology , Transplants , Veins/metabolism , Animals , Capillary Permeability , Disease Models, Animal , Intravital Microscopy/methods , Mice , Neovascularization, Physiologic , Plaque, Atherosclerotic/surgery , Time-Lapse Imaging , Veins/transplantation
6.
Methods ; 128: 52-64, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28669866

ABSTRACT

Intravital microscopy (IVM) is increasingly used in biomedical research to study dynamic processes at cellular and subcellular resolution in their natural environment. Long-term IVM especially can be applied to visualize migration and proliferation over days to months within the same animal without recurrent surgeries. Skin can be repetitively imaged without surgery. To intermittently visualize cells in other organs, such as liver, mammary gland and brain, different imaging windows including the abdominal imaging window (AIW), dermal imaging window (DIW) and cranial imaging window (CIW) have been developed. In this review, we describe the procedure of window implantation and pros and cons of each technique as well as methods to retrace a position of interest over time. In addition, different fluorescent biosensors to facilitate the tracking of cells for different purposes, such as monitoring cell migration and proliferation, are discussed. Finally, we consider new techniques and possibilities of how long-term IVM can be even further improved in the future.


Subject(s)
Fluorescent Dyes , Intravital Microscopy/methods , Microscopy, Fluorescence, Multiphoton/methods , Skin/diagnostic imaging , Animals , Cell Movement/physiology , Humans , Neoplasms/diagnostic imaging , Time Factors
7.
Proc Natl Acad Sci U S A ; 109(48): 19739-44, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-23150545

ABSTRACT

Recent work has demonstrated that following the clearance of infection a stable population of memory T cells remains present in peripheral organs and contributes to the control of secondary infections. However, little is known about how tissue-resident memory T cells behave in situ and how they encounter newly infected target cells. Here we demonstrate that antigen-specific CD8(+) T cells that remain in skin following herpes simplex virus infection show a steady-state crawling behavior in between keratinocytes. Spatially explicit simulations of the migration of these tissue-resident memory T cells indicate that the migratory dendritic behavior of these cells allows the detection of antigen-expressing target cells in physiologically relevant time frames of minutes to hours. Furthermore, we provide direct evidence for the identification of rare antigen-expressing epithelial cells by skin-patrolling memory T cells in vivo. These data demonstrate the existence of skin patrol by memory T cells and reveal the value of this patrol in the rapid detection of renewed infections at a previously infected site.


Subject(s)
Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Epithelium/immunology , Humans
8.
Stem Cells ; 31(3): 602-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23225641

ABSTRACT

It is widely debated whether all tumor cells in mammary tumors have the same potential to propagate and maintain tumor growth or whether there is a hierarchical organization. Evidence for the latter theory is mainly based on the ability or failure of transplanted tumor cells to produce detectable tumors in mice with compromised immune systems; however, this assay has lately been disputed to accurately reflect cell behavior in unperturbed tumors. Lineage tracing experiments have recently shown the existence of a small population of cells, referred to as cancer stem cells (CSCs), that maintains and provides growth of squamous skin tumors and intestinal adenomas. However, the lineage tracing techniques used in these studies provide static images and lack the ability to study whether stem cell properties can be obtained or lost, a process referred to as stem cell plasticity. Here, by intravital lineage tracing, we report for the first time the existence of CSCs in unperturbed mammary tumors and demonstrate CSC plasticity. Our data indicate that existing CSCs disappear and new CSCs form during mammary tumor growth, illustrating the dynamic nature of these cells.


Subject(s)
Mammary Neoplasms, Experimental/pathology , Neoplastic Stem Cells/pathology , Animals , Disease Models, Animal , Female , Mice , Microscopy, Fluorescence, Multiphoton/methods
9.
J Cell Sci ; 124(Pt 3): 299-310, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21242309

ABSTRACT

Metastasis, the process by which cells spread from the primary tumor to a distant site to form secondary tumors, is still not fully understood. Although histological techniques have provided important information, they give only a static image and thus compromise interpretation of this dynamic process. New advances in intravital microscopy (IVM), such as two-photon microscopy, imaging chambers, and multicolor and fluorescent resonance energy transfer imaging, have recently been used to visualize the behavior of single metastasizing cells at subcellular resolution over several days, yielding new and unexpected insights into this process. For example, IVM studies showed that tumor cells can switch between multiple invasion strategies in response to various densities of extracellular matrix. Moreover, other IVM studies showed that tumor cell migration and blood entry take place not only at the invasive front, but also within the tumor mass at tumor-associated vessels that lack an intact basement membrane. In this Commentary, we will give an overview of the recent advances in high-resolution IVM techniques and discuss some of the latest insights in the metastasis field obtained with IVM.


Subject(s)
Microscopy, Fluorescence, Multiphoton/methods , Microscopy, Video/methods , Neoplasm Metastasis/pathology , Neoplasms/pathology , Basement Membrane/metabolism , Basement Membrane/pathology , Cell Movement , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Humans , Microscopy, Video/instrumentation , Neoplasms/blood supply , Neoplasms/metabolism , Neovascularization, Pathologic
10.
EMBO Rep ; 12(9): 931-7, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21760611

ABSTRACT

The death receptor CD95 promotes apoptosis through well-defined signalling pathways. In colorectal cancer cells, CD95 primarily stimulates migration and invasion through pathways that are incompletely understood. Here, we identify a new CD95-activated tyrosine kinase pathway that is essential for CD95-stimulated tumour cell invasion. We show that CD95 promotes Tyr 783 phosphorylation of phospholipase C-γ1 through the platelet-derived growth factor receptor-ß, resulting in ligand-stimulated phosphatidylinositol (4,5)-bisphosphate (PIP(2)) hydrolysis. PIP(2) hydrolysis liberates the actin-severing protein cofilin from the plasma membrane to initiate cortical actin remodelling. Cofilin activation is required for CD95-stimulated formation of membrane protrusions and increased tumour cell invasion.


Subject(s)
Actin Depolymerizing Factors/metabolism , Colorectal Neoplasms/metabolism , Phosphatidylinositols/metabolism , Signal Transduction , fas Receptor/metabolism , Actins/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Membrane/metabolism , Cell Surface Extensions , Colorectal Neoplasms/pathology , Mice , Neoplasm Invasiveness , Phospholipase C gamma/metabolism , Phosphorylation , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptors, Death Domain/metabolism
11.
Front Oncol ; 12: 797453, 2022.
Article in English | MEDLINE | ID: mdl-35756604

ABSTRACT

Cervical cancer is the fourth most common cancer in women worldwide. Squamous cell carcinoma (SCC) and adenocarcinoma (AC) are the most common histological types, with AC patients having worse prognosis. Over the last two decades, incidence rates of AC have increased, highlighting the importance of further understanding AC tumorigenesis, and the need to investigate new treatment options. The cytokine TGF-ß functions as a tumour suppressor in healthy tissue. However, in tumour cells this suppressive function can be overcome. Therefore there is an increasing interest in using TGF-ß inhibitors in the treatment of cancer. Here, we hypothesize that TGF-ß plays a different role in SCC and AC. Analysis of RNA-seq data from the TCGA, using a TGF-ß response signature, resulted in separate clustering of the two subtypes. We further investigated the expression of TGF-ß-signalling related proteins (TßR1/2, SMAD4, pSMAD2, PAI-1, αvß6 and MMP2/9) in a cohort of 62 AC patients. Low TßR2 and SMAD4 expression was associated with worse survival in AC patients and interestingly, high PAI-1 and αvß6 expression was also correlated with worse survival. Similar correlations of TßR2, PAI-1 and αvß6 with clinical parameters were found in previously reported SCC analyses. However, when comparing expression levels between SCC and AC patient samples, pSMAD2, SMAD4, PAI-1 and αvß6 showed lower expression in AC compared to SCC. Because of the low expression of core TßR1/2, (p-)SMAD2 and SMAD4 proteins and the correlation with worse prognosis, TGF-ß pathway most likely leads to tumour inhibitory effects in AC and therefore the use of TGF-ß inhibitors would not be recommended. However, given the correlation of PAI-1 and αvß6 with poor prognosis, the use of TGF- ß inhibitors might be of interest in SCC and in the subsets of AC patients with high expression of these TGF-ß associated proteins.

12.
Methods Mol Biol ; 2488: 47-65, 2022.
Article in English | MEDLINE | ID: mdl-35347682

ABSTRACT

Epithelial to mesenchymal transition (EMT) is crucial during embryonic development, tissue fibrosis, and cancer progression. Epithelial cells that display a cobblestone-like morphology can undergo a switch to mesenchymal-like phenotype, displaying an elongated spindle shape or a fibroblast-like morphology. EMT is characterized by timely and reversible alterations of molecular and cellular processes. The changes include loss of epithelial and gain of mesenchymal marker expression, loss of polarity, increased cell migratory and invasive properties. Epithelial cells can progress unevenly during this transition and attain hybrid E/M states or metastable EMT states, referred to as epithelial cell plasticity. To gain a deeper insight into the mechanism of EMT, understanding the dynamic aspects of this process is essential. One of the most prominent factors to induce EMT is the cytokine transforming growth factor-ß (TGF-ß). This chapter discusses molecular and cellular techniques to monitor TGF-ß-induced signaling and EMT changes in normal and cancer cell lines. These methods include measuring the TGF-ß-induced activation of its intracellular SMAD effectors proteins and changes in epithelial/mesenchymal marker expression and localization. Moreover, we describe assays of cell migration and dynamic reorganization of the actin cytoskeleton and stress filaments that are frequently part of the TGF-ß-induced EMT cellular response.


Subject(s)
Epithelial-Mesenchymal Transition , Transforming Growth Factor beta , Epithelial Cells , Epithelial-Mesenchymal Transition/genetics , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
13.
Cancers (Basel) ; 14(10)2022 May 19.
Article in English | MEDLINE | ID: mdl-35626109

ABSTRACT

Transforming growth factor-ß (TGF-ß) signaling is tightly controlled in duration and intensity during embryonic development and in the adult to maintain tissue homeostasis. To visualize the TGF-ß/SMAD3 signaling kinetics, we developed a dynamic TGF-ß/SMAD3 transcriptional fluorescent reporter using multimerized SMAD3/4 binding elements driving the expression of a quickly folded and highly unstable GFP protein. We demonstrate the specificity and sensitivity of this reporter and its wide application to monitor dynamic TGF-ß/SMAD3 transcriptional responses in both 2D and 3D systems in vitro, as well as in vivo, using live-cell and intravital imaging. Using this reporter in B16F10 cells, we observed single cell heterogeneity in response to TGF-ß challenge, which can be categorized into early, late, and non-responders. Because of its broad application potential, this reporter allows for new discoveries into how TGF-ß/SMAD3-dependent transcriptional dynamics are affected during multistep and reversible biological processes.

14.
Mol Cancer Res ; 20(10): 1516-1531, 2022 10 04.
Article in English | MEDLINE | ID: mdl-35731212

ABSTRACT

Patients with bladder cancer often have a poor prognosis due to the highly invasive and metastatic characteristics of bladder cancer cells. Epithelial-to-mesenchymal transition (EMT) has been causally linked to bladder cancer invasion. The E3 ubiquitin ligase, tumor necrosis factor receptor-associated factor 4 (TRAF4) has been implicated as a tumor promoter in a wide range of cancers. In contrast, here we show that low TRAF4 expression is associated with poor overall survival in patients with bladder cancer. We show that the TRAF4 gene is epigenetically silenced and that ERK mediates TRAF4 phosphorylation, resulting in lower TRAF4 protein levels in bladder cancer cells. In addition, we demonstrate that TRAF4 is inversely correlated with an EMT gene signature/protein marker expression. Functionally, by manipulating TRAF4 expression, we show that TRAF4 regulates EMT genes and epithelial and invasive properties in bladder cancer cells. Transcriptomic analysis of dysregulated TRAF4 expression in bladder cancer cell lines revealed that high TRAF4 expression enhances the bone morphogenetic protein (BMP)/SMAD and inhibits the NF-κB signaling pathway. Mechanistically, we show that TRAF4 targets the E3 ubiquitin ligase SMURF1, a negative regulator of BMP/SMAD signaling, for proteasomal degradation in bladder cancer cells. This was corroborated in patient samples where TRAF4 positively correlates with phospho-SMAD1/5, and negatively correlates with phospho-NFκb-p65. Lastly, we show that genetic and pharmacologic inhibition of SMURF1 inhibits the migration of aggressive mesenchymal bladder cancer cells. IMPLICATIONS: Our findings identify E3 ubiquitin ligase TRAF4 as a potential therapeutic target or biomarker for bladder cancer progression.


Subject(s)
TNF Receptor-Associated Factor 4 , Urinary Bladder Neoplasms , Bone Morphogenetic Proteins/metabolism , Carcinogens , Humans , NF-kappa B/metabolism , Signal Transduction , TNF Receptor-Associated Factor 4/genetics , TNF Receptor-Associated Factor 4/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Urinary Bladder Neoplasms/genetics
15.
Curr Protoc ; 1(5): e116, 2021 May.
Article in English | MEDLINE | ID: mdl-33961349

ABSTRACT

The liver is a frequent site of cancer metastasis, but current treatment options for cancer patients with liver metastasis are limited, resulting in poor prognosis. Colonization of the liver by cancer cells is a multistep and temporally controlled process. Investigating this process in biological relevant settings in a dynamic manner may lead to new therapeutic avenues. Experimental mouse models of liver metastasis combined with high-resolution microscopy methods can facilitate study of the mechanisms that underlie the outgrowth of cancer cells in the liver. Intravital imaging can provide information on the behavior of tumor cells in their biological setting, in time frames of hours to days. In this unit, we describe the experimental induction of liver metastasis through administration of cancer cells into mice via mesenteric vein injection. The behavior of these injected cells can then be studied using intravital imaging by surgical exposure or through an abdominal imaging window. The approach is described for use with an upright multiphoton microscope, making it widely applicable. © 2021 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Inducing liver metastasis through mesenteric vein injection Basic Protocol 2: Short-term imaging of tumor cells in mouse liver Basic Protocol 3: Long-term imaging of tumor cells in mouse liver using an abdominal imaging window.


Subject(s)
Intravital Microscopy , Liver Neoplasms , Animals , Diagnostic Imaging , Disease Models, Animal , Humans , Mice
16.
NPJ Breast Cancer ; 7(1): 140, 2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34707097

ABSTRACT

Reactivation of dormant cancer cells can lead to cancer relapse, metastasis, and patient death. Dormancy is a nonproliferative state and is linked to late relapse and death. No targeted therapy is currently available to eliminate dormant cells, highlighting the need for a deeper understanding and reliable models. Here, we thoroughly characterize the dormant D2.OR and ZR-75-1, and proliferative D2A1 breast cancer cell line models in vivo and/or in vitro, and assess if there is overlap between a dormant and a senescent phenotype. We show that D2.OR but not D2A1 cells become dormant in the liver of an immunocompetent model. In vitro, we show that D2.OR and ZR-75-1 cells in response to a 3D environment or serum-free conditions are growth-arrested in G1, of which a subpopulation resides in a 4NG1 state. The dormancy state is reversible and not associated with a senescence phenotype. This will aid future research on breast cancer dormancy.

17.
Clin Transl Med ; 10(7): e160, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33252863

ABSTRACT

The presence of liver metastases drastically worsens the prognosis of cancer patients. The liver is the second most prevalent metastatic site in cancer patients, but systemic therapeutic opportunities that target liver metastases are still limited. To aid the discovery of novel treatment options for metastatic liver disease, we provide insight into the cellular and molecular steps required for liver colonization. For successful colonization in the liver, adaptation of tumor cells and surrounding stroma is essential. This includes the formation of a pre-metastatic niche, the creation of a fibrotic and immune suppressive environment, angiogenesis, and adaptation of tumor cells. We illustrate that transforming growth factor ß (TGF-ß) is a central cytokine in all these processes. At last, we devise that future research should focus on TGF-ß inhibitory strategies, especially in combination with immunotherapy. This promising systemic treatment strategy has potential to eliminate distant metastases as the efficacy of immunotherapy will be enhanced.

18.
J Control Release ; 320: 19-31, 2020 04 10.
Article in English | MEDLINE | ID: mdl-31899272

ABSTRACT

In cancer treatment, nanomedicines may be employed in an attempt to improve the tumor localization of antineoplastic drugs e.g. immunotherapeutic agents either through passive or active targeting, thereby potentially enhancing therapeutic effect and reducing undesired off-target effects. However, a large number of administrated nanocarriers often fail to reach the tumor area. In the present study, we show that photodynamic therapy (PDT) enhances the tumor accumulation of systemically administered lipid-PEG layer coated poly (lactic-co-glycolic acid) (PLGA) nanoparticles (NP). Intravital microscopy and histological analysis of the tumor area reveal that the tumor vasculature was disrupted after PDT, disturbing blood flow and coinciding with entrapment of nanocarriers in the tumor area. We observed that the nanoparticles accumulating after treatment do not confine to specific locations within the tumor, but rather localize to various cells present throughout the tumor area. Finally, we show by flow cytometry that NP accumulation occurred mostly in immune cells of the myeloid lineage present in the tumor microenvironment (TME) as well as in tumor cells, albeit to a lower extent. These data expose opportunities for combination treatments of clinical PDT with NP-based immunotherapy to modulate the TME and improve antitumor immune responses.


Subject(s)
Antineoplastic Agents , Nanoparticles , Neoplasms , Photochemotherapy , Cell Line, Tumor , Myeloid Cells , Neoplasms/drug therapy
19.
Trends Cancer ; 5(1): 66-78, 2019 01.
Article in English | MEDLINE | ID: mdl-30616757

ABSTRACT

Individual cancer cells can switch, reversibly, to a non-proliferative dormant state, a process characterized by two principal stages: (i) establishment and maintenance, and (ii) the breaking of dormancy. This phenomenon is of clinical importance because dormant cells resist chemotherapy, and this can result in cancer relapse following years, if not decades, of clinical remission. Although the molecular mechanisms governing tumor cell dormancy have not been clearly delineated, accumulating evidence suggests that members of the transforming growth factor-ß (TGF-ß) family are integral. We summarize here recent findings which support the view that TGF-ß family signaling pathways play a pivotal role in cellular dormancy, and discuss how affected cells could be therapeutically targeted to prevent cancer relapse.


Subject(s)
Cell Cycle Checkpoints , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Biomarkers , Epithelial-Mesenchymal Transition/genetics , Humans , Matrix Metalloproteinases/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
20.
Stem Cell Reports ; 10(3): 751-765, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29503086

ABSTRACT

Human pluripotent stem cell (hPSC)-derived kidney organoids may facilitate disease modeling and the generation of tissue for renal replacement. Long-term application, however, will require transferability between hPSC lines and significant improvements in organ maturation. A key question is whether time or a patent vasculature is required for ongoing morphogenesis. Here, we show that hPSC-derived kidney organoids, derived in fully defined medium conditions and in the absence of any exogenous vascular endothelial growth factor, develop host-derived vascularization. In vivo imaging of organoids under the kidney capsule confirms functional glomerular perfusion as well as connection to pre-existing vascular networks in the organoids. Wide-field electron microscopy demonstrates that transplantation results in formation of a glomerular basement membrane, fenestrated endothelial cells, and podocyte foot processes. Furthermore, compared with non-transplanted organoids, polarization and segmental specialization of tubular epithelium are observed. These data demonstrate that functional vascularization is required for progressive morphogenesis of human kidney organoids.


Subject(s)
Kidney Glomerulus/physiology , Kidney Tubules/physiology , Organoids/physiology , Pluripotent Stem Cells/physiology , Animals , Cell Differentiation/physiology , Endothelial Cells/physiology , Humans , Kidney Transplantation/methods , Mice , Morphogenesis/physiology , Podocytes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL