Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Oncol Rep ; 52(4)2024 Oct.
Article in English | MEDLINE | ID: mdl-39092576

ABSTRACT

Senescent cells are known to secrete proteins, including inflammatory cytokines and damage­associated molecular patterns. This phenomenon is known as the senescence­associated secretory phenotype (SASP). SASP in cancer stromal fibroblasts is involved in cancer growth and progression. Conversely, metformin, an antidiabetic drug, has been reported to inhibit SASP induction by inhibiting the activation of NF­κB, a regulator of SASP. To date, at least to the best of our knowledge, there have been no reports regarding cellular senescence in fibroblasts and tumor progression via the SASP­mediated paracrine pathway. The present study thus aimed to elucidate the induction mechanisms of SASP in radiation­induced fibroblasts and to determine its effects on cancer progression via the paracrine pathway. Furthermore, the present study aimed to determine whether controlling SASP using metformin suppresses cancer progression. A well­differentiated esophageal cancer cell line established by the authors' department and fibroblasts isolated and cultured from the non­cancerous esophageal mucosa of resected esophageal cancer cases were used for the experiments. Fibroblasts were irradiated with 8 Gy radiation, and the changes in the expression of the senescence markers, SA­ß­gal, p21, p16 and NF­κB were evaluated using immunofluorescent staining and western blot analysis in the presence or absence of metformin treatment. The culture supernatants of irradiated fibroblasts treated with metformin and those treated without metformin were collected and added to the cancer cells to evaluate their proliferative, invasive and migratory abilities. Vimentin and E­cadherin expression levels were also evaluated using immunofluorescent staining and western blot analysis. The expression levels of p16, p21 and NF­κB in irradiated fibroblasts were attenuated by treatment with metformin. Supernatants collected from irradiated fibroblasts exhibited the proliferative activity of esophageal cancer cells, and the promotion of migratory and invasion abilities, which may be due to epithelial­mesenchymal transition and changes in cell morphology. These reactions were confirmed to be suppressed by the addition of the supernatant of cultured fibroblasts pre­treated with metformin. On the whole, the present study demonstrates that fibroblasts in the cancer stroma may be involved in tumor progression through cellular senescence.


Subject(s)
Cancer-Associated Fibroblasts , Cell Proliferation , Cellular Senescence , Esophageal Neoplasms , Metformin , Metformin/pharmacology , Humans , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Esophageal Neoplasms/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/drug therapy , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/radiation effects , Cancer-Associated Fibroblasts/pathology , Cell Proliferation/drug effects , Disease Progression , NF-kappa B/metabolism , Cell Line, Tumor , Senescence-Associated Secretory Phenotype , Cell Movement/drug effects , Cell Movement/radiation effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/radiation effects , Hypoglycemic Agents/pharmacology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Fibroblasts/drug effects
2.
Cell Rep ; 42(10): 113270, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37851572

ABSTRACT

Esophageal squamous-cell carcinoma (ESCC) is commonly treated with radiotherapy; however, radioresistance hinders its clinical effectiveness, and the underlying mechanism remains elusive. Here, we develop patient-derived xenografts (PDXs) from 19 patients with ESCC to investigate the mechanisms driving radioresistance. Using RNA sequencing, cytokine arrays, and single-cell RNA sequencing, we reveal an enrichment of cancer-associated fibroblast (CAF)-derived collagen type 1 (Col1) and tumor-cell-derived CXCL1 in non-responsive PDXs. Col1 not only promotes radioresistance by augmenting DNA repair capacity but also induces CXCL1 secretion in tumor cells. Additionally, CXCL1 further activates CAFs via the CXCR2-STAT3 pathway, establishing a positive feedback loop. Directly interfering with tumor-cell-derived CXCL1 or inhibiting the CXCL1-CXCR2 pathway effectively restores the radiosensitivity of radioresistant xenografts in vivo. Collectively, our study provides a comprehensive understanding of the molecular mechanisms underlying radioresistance and identifies potential targets to improve the efficacy of radiotherapy for ESCC.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Squamous Cell , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Radiation Tolerance , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/radiation effects , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/radiotherapy , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Chemokine CXCL1/metabolism , Collagen/metabolism , Esophageal Neoplasms/genetics , Esophageal Neoplasms/radiotherapy , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/radiotherapy , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism
3.
Oncol Rep ; 47(5)2022 May.
Article in English | MEDLINE | ID: mdl-35417035

ABSTRACT

Irradiation with a specific wavelength of light using light­emitting diodes (LEDs) has various effects on cells and organisms. Recently, the antitumor effects of visible blue light on tumor cells were reported; however, the mechanism and effects on the tumor microenvironment remain unclear. Human colon cancer cells (HCT­116) were injected into the rectal wall of nude mice. Tumors were irradiated with a 465­nm LED light at 30 mW/cm2 for 30 min. Tumor volumes and the expression levels of opsin 3 (Opn3), autophagy­related factors, cancer­associated fibroblast (CAF) markers, and programmed cell death 1­ligand (PD­L1) were measured. Additionally, human intestinal fibroblasts were cultured in HCT116­conditioned medium (CM) to prepare CAFs. CAFs were divided into an LED group and a control group, and the effect of the LED light on CAF activation in colon cancer cells was examined. Irradiation with blue LED light suppressed tumor growth; Opn3 expression was localized to the cell membrane in the LED group. Irradiated tumors exhibited increased autophagy­related gene expression. Furthermore, in the LED group, TGF­ß and α­SMA expression levels in the fibroblasts were decreased. Regarding CAFs, α­SMA and IL­6 expression levels were decreased in the LED group. HCT­116 cells cultured in CAF­CM with LED irradiation showed no enhanced migration or invasion. In the HCT­116 cells cultured in CM of CAFs irradiated with LED, the relative increase in PD­L1 expression was lower than that noted in the CAF­CM without LED irradiation. Blue LED light may have a direct antitumor effect on colon cancer and also an inhibitory effect on CAFs.


Subject(s)
Cancer-Associated Fibroblasts , Colonic Neoplasms , Light , Animals , B7-H1 Antigen/metabolism , Cancer-Associated Fibroblasts/radiation effects , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Colonic Neoplasms/genetics , HCT116 Cells , Humans , Mice , Mice, Nude , Rod Opsins/metabolism , Tumor Microenvironment
4.
Br J Cancer ; 126(4): 628-639, 2022 03.
Article in English | MEDLINE | ID: mdl-34824448

ABSTRACT

BACKGROUND: The treatment of pancreatic cancer (PDAC) remains clinically challenging, and neoadjuvant therapy (NAT) offers down staging and improved surgical resectability. Abundant fibrous stroma is involved in malignant characteristic of PDAC. We aimed to investigate tissue remodelling, particularly the alteration of the collagen architecture of the PDAC microenvironment by NAT. METHODS: We analysed the alteration of collagen and gene expression profiles in PDAC tissues after NAT. Additionally, we examined the biological role of Ephrin-A5 using primary cultured cancer-associated fibroblasts (CAFs). RESULTS: The expression of type I, III, IV, and V collagen was reduced in PDAC tissues after effective NAT. The bioinformatics approach provided comprehensive insights into NAT-induced matrix remodelling, which showed Ephrin-A signalling as a likely pathway and Ephrin-A5 (encoded by EFNA5) as a crucial ligand. Effective NAT reduced the number of Ephrin-A5+ cells, which were mainly CAFs; this inversely correlated with the clinical tumour shrinkage rate. Experimental exposure to radiation and chemotherapeutic agents suppressed proliferation, EFNA5 expression, and collagen synthesis in CAFs. Forced EFNA5 expression altered CAF collagen gene profiles similar to those found in PDAC tissues after NAT. CONCLUSION: These results suggest that effective NAT changes the extracellular matrix with collagen profiles through CAFs and their Ephrin-A5 expression.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinoma, Pancreatic Ductal/therapy , Collagen/genetics , Ephrin-A5/genetics , Pancreatic Neoplasms/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/radiation effects , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Collagen/metabolism , Ephrin-A5/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Neoadjuvant Therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy , Primary Cell Culture , Retrospective Studies , Signal Transduction , Tumor Cells, Cultured , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects
5.
Front Immunol ; 12: 662594, 2021.
Article in English | MEDLINE | ID: mdl-34177901

ABSTRACT

Cancer-associated fibroblasts (CAFs) participate actively in tumor development and affect treatment responses, by among other mechanisms, promoting an immunosuppressive tumor microenvironment. In contrast to normal fibroblasts, reactive CAFs secrete a myriad of immunomodulatory soluble factors at high levels, i.e. growth factors, cytokines, and chemokines, which directly influence tumor immunity and inflammation. CAFs have been identified as important players in tumor radioresistance. However, knowledge on the immunomodulatory functions of CAFs during/after radiotherapy is still lacking. In this study, we investigated the effects of ionizing radiation on CAF-mediated regulation of dendritic cells (DCs). CAFs were obtained from freshly operated lung cancer tissues, while DCs were procured from peripheral blood of healthy donors. Experimental settings comprised both co-cultures and incubations with conditioned medium from control and irradiated CAFs. Functional assays to study DC differentiation/activation consisted on cytokine release, expression of cell-surface markers, antigen uptake, migration rates, T cell priming, and DC-signaling analysis. We demonstrate that CAFs induce a tolerogenic phenotype in DCs by promoting down-regulation of: i) signature DC markers (CD14, CD1a, CD209); ii) activation markers (CD80, CD86, CD40, and HLA-DR) and iii) functional properties (migration, antigen uptake, and CD4+ T cell priming). Notably, some of these effects were lost in conditioned medium from CAFs irradiated at fractionated medium-dose regimens (3x6 Gy). However, the expression of relevant CAF-derived regulatory agents like thymic stromal lymphopoietin (TSLP) or tryptophan 2,3-dioxygenase (TDO2) was unchanged upon irradiation. This study demonstrates that CAFs interfere with DC immune functions and unveil that certain radiation regimens may reverse CAF-mediated immunosuppressive effects.


Subject(s)
Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/radiation effects , Dendritic Cells/immunology , Immune Tolerance/radiation effects , Radiation, Ionizing , Cell Differentiation/immunology , Coculture Techniques , Dendritic Cells/physiology , Female , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Male , Signal Transduction/immunology
6.
J Radiat Res ; 62(3): 401-413, 2021 May 12.
Article in English | MEDLINE | ID: mdl-33899109

ABSTRACT

Reciprocal communication between the malignant and non-malignant cellular elements in tumors is essential for cancer sustainability and plays an important role in the response of cancers to treatments. Some of this cellular crosstalk takes place via secretion of vesicles that are actively released into the extracellular space by most cell types in tumors. Recent studies have demonstrated radiation-induced changes in the secretion rate and composition of extracellular vesicles (EVs), with impact on radiation-related cellular communication. However, little is known about the effects of different radiation regimens on the release of EVs by cells of the tumor microenvironment. In this study, we provide a comprehensive molecular characterization of EVs released by cultured primary lung tumor fibroblasts. We explore the quantitative and morphological changes triggered by ionizing radiation (IR), delivered as a single dose of 18 Gy or three consecutive daily medium-doses of 6 Gy. Cancer-associated fibroblasts (CAFs) secrete EVs with sizes ranging from 80 to 200 nm, expressing some of the classical exosome markers. Exposing CAFs to a single-high radiation dose (1 × 18 Gy) or fractionated medium-dose did not alter the release of CAF-EVs. The protein composition of CAF-EVs was analyzed by LC-MS/MS proteomics and revealed that CAF-EVs are enriched with heat shock proteins, integrins, tetraspanins, proteinases, collagens, growth factors and an array of molecules involved in the regulation of cell migration and the immune system. Quantitative proteomic analyses revealed minor changes in the protein composition of CAF-EVs after radiation exposure. Taken together, this study presents original data on lung tumor CAF-EV composition and reveals that release and protein cargo of CAF-EVs are largely unaltered after exposing CAFs to IR.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/radiation effects , Extracellular Vesicles/metabolism , Extracellular Vesicles/radiation effects , Proteins/metabolism , Radiation, Ionizing , Apoptosis/radiation effects , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cellular Senescence/radiation effects , Extracellular Vesicles/ultrastructure , Female , Humans , Male
7.
Curr Radiopharm ; 14(4): 374-393, 2021.
Article in English | MEDLINE | ID: mdl-33357190

ABSTRACT

The tumor microenvironment is a dynamic ecosystem where malignant cells interact with the stromal cells sustaining and promoting tumor growth and metastasis. Cancer-associated fibroblasts (CAFs) are the major component of tumor stroma. CAFs control key tumorigenic activities by participating in immune evasion and suppression, extracellular matrix remodeling, neo-angiogenesis, and drug resistance. Therefore, targeting CAFs emerges as an attractive anti-cancer strategy. This review summarized recent advancements in targeting CAFs with diagnostic and therapeutic radiopharmaceuticals using clinically-promising biomarkers. The efforts to improve clinical outcomes via the application of new radiotheranostic compounds are discussed in the context of radionuclide, the pharmacophore, and, more generally, in terms of biomarker specificity and expression across different cancers and CAF phenotypes.


Subject(s)
Cancer-Associated Fibroblasts/radiation effects , Precision Medicine , Radiopharmaceuticals/pharmacology , Tumor Microenvironment , Cancer-Associated Fibroblasts/physiology , Humans , Phenotype
8.
Sci Rep ; 10(1): 12096, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32694592

ABSTRACT

One of the major issues in cancer radiotherapy (RT) is normal tissue toxicity. Introduction of radiosensitizers like gold nanoparticles (GNPs) into cancer cells to enhance the local RT dose has been tested successfully. However, it is not known how GNPs interact with other stromal cells such as normal fibroblasts (FBs) and cancer associated fibroblasts (CAFs) within the tumour microenvironment. It is known that FBs turn into CAFs to promote tumour growth. Hence, we used FBs and CAFs along with HeLa (our cancer cell line) to evaluate the differences in GNP uptake and resulting radiation induced damage to elucidate the GNP-mediated therapeutic effect in RT. The CAFs had the largest uptake of the GNPs per cell, with on average 265% relative to HeLa while FBs had only 7.55% the uptake of HeLa and 2.87% the uptake of CAFs. This translated to increases in 53BP1-related DNA damage foci in CAFs (13.5%) and HeLa (9.8%) compared to FBs (8.8%) with RT treatment. This difference in DNA damage due to selective targeting of cancer associated cells over normal cells may allow GNPs to be an effective tool in future cancer RT to battle normal tissue toxicity while improving local RT dose to the tumour.


Subject(s)
Gold/pharmacology , Neoplasms/metabolism , Radiation-Sensitizing Agents/pharmacology , Tumor Suppressor p53-Binding Protein 1/metabolism , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Damage , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Gold/chemistry , HeLa Cells , Humans , Metal Nanoparticles , Neoplasms/therapy , Radiation-Sensitizing Agents/chemistry , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects
9.
Mol Carcinog ; 59(7): 754-765, 2020 07.
Article in English | MEDLINE | ID: mdl-32363633

ABSTRACT

Fibroblasts are a key component of the tumor microenvironment (TME) that can serve as a scaffold for tumor cell migration and augment the tumor's ability to withstand harsh conditions. When activated by external or endogenous stimuli, normal fibroblasts become cancer associated fibroblasts (CAFs), a heterogeneous group of stromal cells in the tumor that are phenotypically and epigenetically different from normal fibroblasts. Dynamic crosstalk between cancer cells, immune cells, and CAFs through chemokines and surface signaling makes the TME conducive to tumor growth. When activated, CAFs promote tumorigenesis and metastasis through several phenomena including regulation of tumor immunity, metabolic reprogramming of the TME, extracellular matrix remodeling and contraction, and induction of therapeutic resistance. Ionizing radiation (radiation theraphy [RT]) is a potent immunological stimulant that has been shown to increase cytotoxic Teff infiltration and IFN-I stimulated genes. RT, however, is unable to overcome the infiltration and activation of immunosuppressive cells which can contribute to tumor progression. Another paradox of RT is that, while very effective at killing cancer cells, it can contribute to the formation of CAFs. This review examines how the interplay between CAFs and immune cells during RT contributes to organ fibrosis, immunosuppression, and tumor growth. We focus on targeting mechanistic pathways of CAF formation as a potentially effective strategy not only for preventing organ fibrosis, but also in hampering tumor progression in response to RT.


Subject(s)
Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/pathology , Neoplasms/immunology , Neoplasms/pathology , Animals , Cancer-Associated Fibroblasts/radiation effects , Fibrosis/immunology , Fibrosis/pathology , Humans , Neoplasms/radiotherapy , Radiotherapy/methods , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
10.
J Exp Clin Cancer Res ; 39(1): 65, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32293494

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAFs) have been intensively studied in recent studies with aims of finding more concrete evidence on their mechanism of involvement in tumor progression, which is currently unknown. CAFs can secrete exosomes which are loaded with proteins, lipids and RNAs, all of which affect tumor microenvironment. The present study identified microRNA-93-5p (miR-93-5p) as a novel exosomal cargo responsible for the pro-tumorigenic effects of CAFs on colorectal cancer (CRC). METHODS: CAFs and normal fibroblasts (NFs) were isolated from cancerous tissues and matched with paracancerous tissues that had been surgically resected from CRC patients. The interaction among miR-93-5p, forkhead box A1 (FOXA1) and TGFB3 was identified through ChIP and dual luciferase reporter assays. The proliferation and apoptosis of SW480 cells co-cultured with CAFs-derived exosomes under irradiation were evaluated by CCK-8, colony formation, and flow cytometric assays. Tumorigenesis of SW480 cells in nude mice was assessed under the irradiation. RESULTS: FOXA1 was found to be associated with reduced radioresistance in CRC cells and was verified as a target of miR-93-5p. CAFs-derived exosomes contained higher miR-93-5p than those from NFs, which augmented SW480 cell proliferation and rescued them from radiation-induced apoptosis. miR-93-5p was identified as a mediator of the exosomal effects of CAFs on SW480 cells, possibly through downregulating FOXA1 and upregulating TGFB3. FOXA1 could bind to the promoter of TGFB3, thereby inhibiting nuclear accumulation of TGFB3. Also, CAFs-derived exosomes containing miR-93-5p increased the tumor growth of SW480 cells in irradiated nude mice. CONCLUSION: The present study identifies miR-93-5p as a specific exosomal cargo that rescues CRC cells against radiation-induced apoptosis.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/radiotherapy , Exosomes/metabolism , Hepatocyte Nuclear Factor 3-alpha/metabolism , MicroRNAs/metabolism , Transforming Growth Factor beta3/metabolism , Aged , Animals , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/radiation effects , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Down-Regulation , Female , HT29 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Middle Aged , Radiation Tolerance , Transforming Growth Factor beta3/genetics , Up-Regulation
11.
Cancer Res ; 80(8): 1681-1692, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32086240

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is highly resistant to radiotherapy, chemotherapy, or a combination of these modalities, and surgery remains the only curative intervention for localized disease. Although cancer-associated fibroblasts (CAF) are abundant in PDAC tumors, the effects of radiotherapy on CAFs and the response of PDAC cells to radiotherapy are unknown. Using patient samples and orthotopic PDAC biological models, we showed that radiotherapy increased inducible nitric oxide synthase (iNOS) in the tumor tissues. Mechanistic in vitro studies showed that, although undetectable in radiotherapy-activated tumor cells, iNOS expression and nitric oxide (NO) secretion were significantly increased in CAFs secretome following radiotherapy. Culture of PDAC cells with conditioned media from radiotherapy-activated CAFs increased iNOS/NO signaling in tumor cells through NF-κB, which, in turn, elevated the release of inflammatory cytokines by the tumor cells. Increased NO after radiotherapy in PDAC contributed to an acidic microenvironment that was detectable using the radiolabeled pH (low) insertion peptide (pHLIP). In murine orthotopic PDAC models, pancreatic tumor growth was delayed when iNOS inhibition was combined with radiotherapy. These data show the important role that iNOS/NO signaling plays in the effectiveness of radiotherapy to treat PDAC tumors. SIGNIFICANCE: A radiolabeled pH-targeted peptide can be used as a PET imaging tool to assess therapy response within PDAC and blocking iNOS/NO signaling may improve radiotherapy outcomes.


Subject(s)
Cancer-Associated Fibroblasts/radiation effects , Carcinoma, Pancreatic Ductal/radiotherapy , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Pancreatic Neoplasms/radiotherapy , Animals , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Culture Media, Conditioned , Cytokines/metabolism , Humans , Mice , NF-kappa B/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Treatment Outcome , Tumor Microenvironment
12.
Int J Mol Sci ; 20(16)2019 Aug 13.
Article in English | MEDLINE | ID: mdl-31412584

ABSTRACT

Melatonin exerts oncostatic actions and sensitizes tumor cells to chemotherapeutics or radiation. In our study, we investigated the effects of docetaxel, vinorelbine, and radiation on human breast fibroblasts and its modulation by melatonin. Docetaxel or vinorelbine inhibits proliferation and stimulates the differentiation of breast preadipocytes, by increasing C/EBPα and PPARγ expression and by downregulating tumor necrosis factor α (TNFα), interleukin 6 (IL-6), and IL-11 expression. Radiation inhibits both proliferation and differentiation through the downregulation of C/EBPα and PPARγ and by stimulating TNFα expression. In addition, docetaxel and radiation decrease aromatase activity and expression by decreasing aromatase promoter II and cyclooxygenases 1 and 2 (COX-1 and COX-2) expression. Melatonin potentiates the stimulatory effect of docetaxel and vinorelbine on differentiation and their inhibitory effects on aromatase activity and expression, by increasing the stimulatory effect on C/EBPα and PPARγ expression and the downregulation of antiadipogenic cytokines and COX expression. Melatonin also counteracts the inhibitory effect of radiation on differentiation of preadipocytes, by increasing C/EBPα and PPARγ expression and by decreasing TNFα expression. Melatonin also potentiates the inhibitory effect exerted by radiation on aromatase activity and expression by increasing the downregulation of promoter II, and COX-1 and COX-2 expression. Our findings suggest that melatonin modulates regulatory effects induced by chemotherapeutic drugs or radiation on preadipocytes, which makes it a promising adjuvant for chemotherapy and radiotherapy sensibilization.


Subject(s)
Antineoplastic Agents/pharmacology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/radiation effects , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , Melatonin/pharmacology , Radiation, Ionizing , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/radiation effects , Aromatase/metabolism , Breast Neoplasms , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cancer-Associated Fibroblasts/metabolism , Docetaxel/pharmacology , Enzyme Activation/drug effects , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Humans , Mammary Glands, Human/cytology , PPAR gamma/genetics , PPAR gamma/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Vinorelbine/pharmacology
13.
Ann Palliat Med ; 8(3): 326-336, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31370663

ABSTRACT

Radiation therapy is commonly used in the metastatic setting to palliate pain, neurological deficits, bleeding and other complications of metastatic disease, allowing patients to live longer and have better quality of life. Despite the effective use of radiation and other palliative treatment modalities, many patients continue to experience poorly controlled pain and other serious sequelae of their disease, underscoring the need for additional research in this area. In this review we highlight recent developments impacting the fields of palliative care and radiation oncology and describe opportunities for research and innovation including studies of tumor microenvironment, identification of effective biomarkers of tumor response and combinatorial treatments with new systemic agents. It is our hope that progress in these fields will improve the lives of patients living with advanced malignancies.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Cancer Pain/radiotherapy , Neoplasms/radiotherapy , Palliative Care/methods , Translational Research, Biomedical/organization & administration , Animals , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/pharmacology , Biomarkers, Tumor , Bone Neoplasms/radiotherapy , Bone Neoplasms/secondary , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Cancer-Associated Fibroblasts/radiation effects , Combined Modality Therapy , Humans , Immune System/radiation effects , Life Expectancy , Neoplasm Metastasis , Neoplasms/pathology , Neoplasms/therapy , Quality of Life , Radiation Oncology , Spinal Neoplasms/radiotherapy , Spinal Neoplasms/secondary , Tumor Microenvironment/radiation effects
14.
Front Immunol ; 10: 193, 2019.
Article in English | MEDLINE | ID: mdl-30828330

ABSTRACT

In recent decades, there has been substantial growth in our understanding of the immune system and its role in tumor growth and overall survival. A central finding has been the cross-talk between tumor cells and the surrounding environment or stroma. This tumor stroma, comprised of various cells, and extracellular matrix (ECM), has been shown to aid in suppressing host immune responses against tumor cells. Through immunosuppressive cytokine secretion, metabolic alterations, and other mechanisms, the tumor stroma provides a complex network of safeguards for tumor proliferation. With recent advances in more effective, localized treatment, radiation therapy (XRT) has allowed for strategies that can effectively alter and ablate tumor stromal tissue. This includes promoting immunogenic cell death through tumor antigen release to increasing immune cell trafficking, XRT has a unique advantage against the tumoral immune evasion mechanisms that are orchestrated by stromal cells. Current studies are underway to elucidate pathways within the tumor stroma as potential targets for immunotherapy and chemoradiation. This review summarizes the effects of tumor stroma in tumor immune evasion, explains how XRT may help overcome these effects, with potential combinatorial approaches for future treatment modalities.


Subject(s)
Neoplasms/pathology , Neoplasms/radiotherapy , Stromal Cells/radiation effects , Tumor Microenvironment/radiation effects , Animals , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/radiation effects , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Extracellular Matrix/radiation effects , Humans , Immunity , Immunomodulation/radiation effects , Neoplasms/immunology , Radiation Tolerance/immunology , Radiation Tolerance/radiation effects , Radiotherapy , Stromal Cells/immunology , Tumor Microenvironment/immunology
15.
Int J Radiat Oncol Biol Phys ; 102(2): 417-425, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30191873

ABSTRACT

PURPOSE: Gorlin syndrome (or basal-cell nevus syndrome) is a cancer-prone genetic disease in which hypersusceptibility to secondary cancer and tissue reaction after radiation therapy is debated, as is increased radiosensitivity at cellular level. Gorlin syndrome results from heterozygous mutations in the PTCH1 gene for 60% of patients, and we therefore aimed to highlight correlations between intrinsic radiosensitivity and PTCH1 gene expression in fibroblasts from adult patients with Gorlin syndrome. METHODS AND MATERIALS: The radiosensitivity of fibroblasts from 6 patients with Gorlin syndrome was determined by cell-survival assay after high (0.5-3.5 Gy) and low (50-250 mGy) γ-ray doses. PTCH1 and DNA damage response gene expression was characterized by real-time polymerase chain reaction and Western blotting. DNA damage and repair were investigated by γH2AX and 53BP1 foci assay. PTCH1 knockdown was performed in cells from healthy donors by using stable RNA interference. Gorlin cells were genotyped by 2 complementary sequencing methods. RESULTS: Only cells from patients with Gorlin syndrome who presented severe deficiency in PATCHED1 protein exhibited a significant increase in cellular radiosensitivity, affecting cell responses to both high and low radiation doses. For 2 of the radiosensitive cell strains, heterozygous mutations in the 5' end of PTCH1 gene explain PATCHED1 protein deficiency. In all sensitive cells, DNA damage response pathways (ATM, CHK2, and P53 levels and activation by phosphorylation) were deregulated after irradiation, whereas DSB repair recognition was unimpaired. Furthermore, normal cells with RNA interference-mediated PTCH1 deficiency showed reduced survival after irradiation, directly linking this gene to high- and low-dose radiosensitivity. CONCLUSIONS: In the present study, we show an inverse correlation between PTCH1 expression level and cellular radiosensitivity, suggesting an explanation for the conflicting results previously reported for Gorlin syndrome and possibly providing a basis for prognostic screens for radiosensitive patients with Gorlin syndrome and PTCH1 mutations.


Subject(s)
Basal Cell Nevus Syndrome/genetics , Cancer-Associated Fibroblasts/radiation effects , Patched-1 Receptor/deficiency , Radiation Tolerance/genetics , Adult , Cell Survival/radiation effects , DNA Damage/genetics , DNA Repair/genetics , Female , Histones/genetics , Humans , Male , Middle Aged , Patched-1 Receptor/genetics , Tumor Suppressor p53-Binding Protein 1/genetics
16.
Cancer Res ; 78(3): 659-670, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29217764

ABSTRACT

Preoperative radiotherapy (RT) is a mainstay in the management of rectal cancer, a tumor characterized by desmoplastic stroma containing cancer-associated fibroblasts (CAF). Although CAFs are abundantly present, the effects of RT to CAF and its impact on cancer cells are unknown. We evaluated the damage responses of CAF to RT and investigated changes in colorectal cancer cell growth, transcriptome, metabolome, and kinome in response to paracrine signals emerging from irradiated CAF. RT to CAF induced DNA damage, p53 activation, cell-cycle arrest, and secretion of paracrine mediators, including insulin-like growth factor-1 (IGF1). Subsequently, RT-activated CAFs promoted survival of colorectal cancer cells, as well as a metabolic switch favoring glutamine consumption through IGF1 receptor (IGF1R) activation. RT followed by IGF1R neutralization in orthotopic colorectal cancer models reduced the number of mice with organ metastases. Activation of the downstream IGF1R mediator mTOR was significantly higher in matched (intrapatient) samples and in unmatched (interpatient) samples from rectal cancer patients after neoadjuvant chemoradiotherapy. Taken together, our data support the notion that paracrine IGF1/IGF1R signaling initiated by RT-activated CAF worsens colorectal cancer progression, establishing a preclinical rationale to target this activation loop to further improve clinical responses and patient survival.Significance: These findings reveal that paracrine IGF1/IGF1R signaling promotes colorectal cancer progression, establishing a preclinical rationale to target this activation loop. Cancer Res; 78(3); 659-70. ©2017 AACR.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/pathology , Gamma Rays/adverse effects , Paracrine Communication , Receptors, Somatomedin/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/radiotherapy , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/radiation effects , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/radiation effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/radiotherapy , Female , Humans , Metabolome , Mice , Mice, Nude , Prognosis , Signal Transduction , Transcriptome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
BMC Cancer ; 17(1): 675, 2017 10 07.
Article in English | MEDLINE | ID: mdl-28987144

ABSTRACT

BACKGROUND: Cutaneous basal cell carcinoma (BCC) is the commonest cancer worldwide. BCC is locally invasive and the surrounding stromal microenvironment is pivotal for tumourigenesis. Cancer associated fibroblasts (CAFs) in the microenvironment are essential for tumour growth in a variety of neoplasms but their role in BCC is poorly understood. METHODS: Material included facial BCC and control skin from the peritumoural area and from the buttocks. With next-generation sequencing (NGS) we compared mRNA expression between BCC and peritumoural skin. qRT-PCR, immunohistochemical and immunofluorescent staining were performed to validate the NGS results and to investigate CAF-related cyto-and chemokines. RESULTS: NGS revealed upregulation of 65 genes in BCC coding for extracellular matrix components pointing at CAF-related matrix remodeling. qRT-PCR showed increased mRNA expression of CAF markers FAP-α, PDGFR-ß and prolyl-4-hydroxylase in BCC. Peritumoural skin (but not buttock skin) also exhibited high expression of PDGFR-ß and prolyl-4-hydroxylase but not FAP-α. We found a similar pattern for the CAF-associated chemokines CCL17, CCL18, CCL22, CCL25, CXCL12 and IL6 with high expression in BCC and peritumoural skin but absence in buttock skin. Immunofluorescence revealed correlation between FAP-α and PDGFR-ß and CXCL12 and CCL17. CONCLUSION: Matrix remodeling is the most prominent molecular feature of BCC. CAFs are present within BCC stroma and associated with increased expression of chemokines involved in tumour progression and immunosuppression (CXCL12, CCL17). Fibroblasts from chronically sun-exposed skin near tumours show gene expression patterns resembling that of CAFs, indicating that stromal fibroblasts in cancer-free surgical BCC margins exhibit a tumour promoting phenotype.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinogenesis/genetics , Carcinoma, Basal Cell/genetics , Skin Neoplasms/genetics , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/radiation effects , Carcinogenesis/radiation effects , Carcinoma, Basal Cell/pathology , Chemokine CCL17/genetics , Chemokine CCL22/genetics , Chemokine CXCL12/genetics , Chemokines, CC/genetics , Gene Expression Regulation, Neoplastic/radiation effects , High-Throughput Nucleotide Sequencing , Humans , Interleukin-6/genetics , RNA, Messenger/genetics , Skin/pathology , Skin/radiation effects , Skin Neoplasms/pathology , Sunlight/adverse effects , Tumor Microenvironment/genetics , Tumor Microenvironment/radiation effects
18.
Future Oncol ; 13(17): 1537-1550, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28685611

ABSTRACT

Solid tumors are composed of tumor epithelial cells and the stroma, which are seemingly separate but actually related through cell-cell and cell-matrix interactions. These interactions can promote tumor evolution. Cancer-associated fibroblasts (CAFs) are the most abundant non-neoplastic cells in the stroma and also among the most important cell types interacting with cancer cells. Particularly, cancer cells promote the formation and maintenance of CAFs by secreting various cytokines. The activated CAFs then synthesize a series of growth factors to promote tumor cell growth, invasion and metastasis. More importantly, the presence of CAFs also interferes with therapeutic efficacy, bringing severe challenges to radiotherapy. This review summarizes the effect of CAFs on the radiosensitivity of tumor cells and underscores the need for further studies on CAFs in order to improve the efficacy of antitumor therapy.


Subject(s)
Cancer-Associated Fibroblasts/radiation effects , Neoplasms/radiotherapy , Radiation Tolerance/genetics , Tumor Microenvironment/genetics , Cancer-Associated Fibroblasts/metabolism , Cell Communication/genetics , Cell Communication/radiation effects , Cell Proliferation/genetics , Humans , Neoplasms/pathology
19.
Sci Rep ; 7: 46714, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28440285

ABSTRACT

Cancer-associated fibroblasts (CAFs) are abundantly present in solid tumors and affect tumorigenesis and therapeutic responses. In the context of clinical radiotherapy, the impact of irradiated CAFs to treatment outcomes is largely unexplored. Aiming at improving radiotherapy efficacy, we have here explored the effect of radiation on the inherent pro-tumorigenic capacity of CAFs in animals. Ionizing radiation was delivered to cultured CAFs as single-high or fractionated doses. Tumor development was compared in mice receiving A549 lung tumor cells admixed with irradiated or control CAFs. Biological mechanisms behind tumor growth regulation were investigated by quantitative histology and immunohistochemistry. Viability assessments confirmed that irradiated CAFs are fully functional prior to implantation. However, the enhanced tumorigenic effect observed in tumors co-implanted with control CAFs was abrogated in tumors established with irradiated CAFs. Experiments to ascertain fate of implanted fibroblasts showed that exogenously administered CAFs reside at the implantation site for few days, suggesting that tumor growth regulation from admixed CAFs take place during initial tumor formation. Our work demonstrate that irradiated CAFs lose their pro-tumorigenic potential in vivo, affecting angiogenesis and tumor engraftment. This finding propose a previously unknown advantageous effect induced by radiotherapy, adding to the direct cytotoxic effects on transformed epithelial cells.


Subject(s)
Cancer-Associated Fibroblasts/radiation effects , Carcinogenesis/pathology , Carcinoma, Squamous Cell/pathology , Lung Neoplasms/pathology , Neovascularization, Pathologic/prevention & control , Animals , Apoptosis , Cancer-Associated Fibroblasts/pathology , Carcinogenesis/radiation effects , Carcinoma, Squamous Cell/radiotherapy , Cell Proliferation , Female , Humans , Lung Neoplasms/radiotherapy , Mice , Mice, Nude , Radiation, Ionizing , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL