1
|
Sui L, Wang J, Jiang WG, Song X, Ye L. Molecular mechanism of bone metastasis in breast cancer. Front Oncol 2024; 14:1401113. [PMID: 39605887 PMCID: PMC11599183 DOI: 10.3389/fonc.2024.1401113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Bone metastasis is a debilitating complication that frequently occurs in the advanced stages of breast cancer. However, the underlying molecular and cellular mechanisms of the bone metastasis remain unclear. Here, we elucidate how bone metastasis arises from tumor cells that detach from the primary lesions and infiltrate into the surrounding tissue, as well as how these cells disseminate to distant sites. Specifically, we elaborate how tumor cells preferentially grow within the bone micro-environment and interact with bone cells to facilitate bone destruction, characterized as osteoclastic bone metastasis, as well as new bone matrix deposition, characterized as osteoblastic bone metastasis. We also updated the current understanding of the molecular mechanisms underlying bone metastasis and reasons for relapse in breast cancer, and also opportunities of developing novel diagnostic approaches and treatment.
Collapse
Affiliation(s)
- Laijian Sui
- Department of Orthopedics, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Jing Wang
- Department of Intensive Care Unit, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Wen G. Jiang
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Xicheng Song
- Department of Otorhinolaryngol and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| |
Collapse
|
2
|
Zhu Y, Neelamegham S. Knockout studies using CD34+ hematopoietic cells suggest that CD44 is a physiological human neutrophil E-selectin ligand. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.18.553923. [PMID: 37645985 PMCID: PMC10462143 DOI: 10.1101/2023.08.18.553923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The recruitment of peripheral blood neutrophils at sites of inflammation involves a multistep cascade, starting with E- and P-selectin expressed on the inflamed vascular endothelium binding sialofucosylated glycans on leukocytes. As the glycoconjugate biosynthesis pathways in different cells are distinct, the precise carbohydrate ligands of selectins varies both across species, and between different immune cell populations in a given species. To study this aspect in human neutrophils, we developed a protocol to perform CRISPR/Cas9 gene-editing on CD34+ hHSCs (human hematopoietic stem/progenitor cells) as they are differentiated towards neutrophil lineage. This protocol initially uses a cocktail of SCF (stem-cell factor), IL-3 (interleukin-3) and FLT-3L (FMS-like tyrosine kinase 3 ligand) to expand the stem/progenitor cells followed by directed differentiation to neutrophils using G-CSF (granulocyte colony-stimulating factor). Microfluidics based assays were performed on a confocal microscope platform to characterize the rolling phenotype of each edited cell type in mixed populations. These studies demonstrated that CD44, but not CD43, is a major E-selectin ligand on human neutrophils. The loss of function results were validated by developing sialofucosylated recombinant CD44. This glycosylated protein supported both robust E-selectin binding in a cell-free assay, and it competitively blocked neutrophil adhesion to E-selectin on inflamed endothelial cells. Together, the study establishes important methods to study human neutrophil biology and determines that sialoflucosylated-CD44 is a physiological human E-selectin ligand.
Collapse
Affiliation(s)
- Yuqi Zhu
- Department of Chemical and Biological Engineering, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260
| | - Sriram Neelamegham
- Department of Chemical and Biological Engineering, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260
- Department of Biomedical Engineering, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260
- Department of Medicine School of Engineering and Applies Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260
| |
Collapse
|
3
|
Mahgoub E, Taneera J, Sulaiman N, Saber-Ayad M. The role of autophagy in colorectal cancer: Impact on pathogenesis and implications in therapy. Front Med (Lausanne) 2022; 9:959348. [PMID: 36160153 PMCID: PMC9490268 DOI: 10.3389/fmed.2022.959348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is considered as a global major cause of cancer death. Surgical resection is the main line of treatment; however, chemo-, radiotherapy and other adjuvant agents are crucial to achieve good outcomes. The tumor microenvironment (TME) is a well-recognized key player in CRC progression, yet the processes linking the cancer cells to its TME are not fully delineated. Autophagy is one of such processes, with a controversial role in the pathogenesis of CRC, with its intricate links to many pathological factors and processes. Autophagy may apparently play conflicting roles in carcinogenesis, but the precise mechanisms determining the overall direction of the process seem to depend on the context. Additionally, it has been established that autophagy has a remarkable effect on the endothelial cells in the TME, the key substrate for angiogenesis that supports tumor metastasis. Favorable response to immunotherapy occurs only in a specific subpopulation of CRC patients, namely the microsatellite instability-high (MSI-H). In view of such limitations of immunotherapy in CRC, modulation of autophagy represents a potential adjuvant strategy to enhance the effect of those relatively safe agents on wider CRC molecular subtypes. In this review, we discussed the molecular control of autophagy in CRC and how autophagy affects different processes and mechanisms that shape the TME. We explored how autophagy contributes to CRC initiation and progression, and how it interacts with tumor immunity, hypoxia, and oxidative stress. The crosstalk between autophagy and the TME in CRC was extensively dissected. Finally, we reported the clinical efforts and challenges in combining autophagy modulators with various cancer-targeted agents to improve CRC patients’ survival and restrain cancer growth.
Collapse
Affiliation(s)
- Eglal Mahgoub
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Jalal Taneera
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Nabil Sulaiman
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Maha Saber-Ayad
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Faculty of Medicine, Cairo University, Giza, Egypt
- *Correspondence: Maha Saber-Ayad,
| |
Collapse
|
4
|
Zou Q, Lei X, Xu A, Li Z, He Q, Huang X, Xu G, Tian F, Ding Y, Zhu W. Chemokines in progression, chemoresistance, diagnosis, and prognosis of colorectal cancer. Front Immunol 2022; 13:724139. [PMID: 35935996 PMCID: PMC9353076 DOI: 10.3389/fimmu.2022.724139] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 06/27/2022] [Indexed: 12/24/2022] Open
Abstract
Plenty of factors affect the oncogenesis and progression of colorectal cancer in the tumor microenvironment, including various immune cells, stromal cells, cytokines, and other factors. Chemokine is a member of the cytokine superfamily. It is an indispensable component in the tumor microenvironment. Chemokines play an antitumor or pro-tumor role by recruitment or polarization of recruiting immune cells. Meanwhile, chemokines, as signal molecules, participate in the formation of a cross talk among signaling pathways and non-coding RNAs, which may be involved in promoting tumor progression. In addition, they also function in immune escape. Chemokines are related to drug resistance of tumor cells and may even provide reference for the diagnosis, therapy, and prognosis of patients with colorectal cancer.
Collapse
Affiliation(s)
- Qian Zou
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Xue Lei
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Aijing Xu
- Department of Genetics and Endocrinology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ziqi Li
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Qinglian He
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Xiujuan Huang
- Department of Pathology, Guangdong Medical University, Dongguan, China
- Department of Hematology, Longgang District People’s Hospital of Shenzhen, Shenzhen, China
| | - Guangxian Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Institute of Clinical Laboratory, Guangdong Medical University, Dongguan, China
| | - Faqing Tian
- Department of Pathology, Guangdong Medical University, Dongguan, China
- Department of Genetics and Endocrinology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Faqing Tian, ; Yuanlin Ding, ; Wei Zhu,
| | - Yuanlin Ding
- School of Public Health, Guangdong Medical University, Dongguan, China
- *Correspondence: Faqing Tian, ; Yuanlin Ding, ; Wei Zhu,
| | - Wei Zhu
- Department of Pathology, Guangdong Medical University, Dongguan, China
- *Correspondence: Faqing Tian, ; Yuanlin Ding, ; Wei Zhu,
| |
Collapse
|
5
|
Makaremi S, Asadzadeh Z, Hemmat N, Baghbanzadeh A, Sgambato A, Ghorbaninezhad F, Safarpour H, Argentiero A, Brunetti O, Bernardini R, Silvestris N, Baradaran B. Immune Checkpoint Inhibitors in Colorectal Cancer: Challenges and Future Prospects. Biomedicines 2021; 9:1075. [PMID: 34572263 PMCID: PMC8467932 DOI: 10.3390/biomedicines9091075] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy is a new pillar of cancer therapy that provides novel opportunities to treat solid tumors. In this context, the development of new drugs targeting immune checkpoints is considered a promising approach in colorectal cancer (CRC) treatment because it can be induce specific and durable anti-cancer effects. Despite many advances in the immunotherapy of CRC, there are still limitations and obstacles to successful treatment. The immunosuppressive function of the tumor microenvironment (TME) is one of the causes of poor response to treatment in CRC patients. For this reason, checkpoint-blocking antibodies have shown promising outcomes in CRC patients by blocking inhibitory immune checkpoints and enhancing immune responses against tumors. This review summarizes recent advances in immune checkpoint inhibitors (ICIs), such as CTLA-4, PD-1, PD-L1, LAG-3, and TIM-3 in CRC, and it discusses various therapeutic strategies with ICIs, including the double blockade of ICIs, combination therapy of ICIs with other immunotherapies, and conventional treatments. This review also delineates a new hopeful path in the combination of anti-PD-1/anti-PD-L1 with other ICIs such as anti-CTLA-4, anti-LAG-3, and anti-TIM-3 for CRC treatment.
Collapse
Affiliation(s)
- Shima Makaremi
- Department of Immunology & Microbiology, School of Medicine, Arak University of Medical Sciences, Arak 3848176941, Iran;
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
| | - Alessandro Sgambato
- Istituto di Ricovero e Cura a Carattere Scientifico Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 5972362 Rome, Italy;
- Area of Pathology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 5972362 Rome, Italy
| | - Farid Ghorbaninezhad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - Hossein Safarpour
- Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
| | - Antonella Argentiero
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (A.A.); (O.B.)
| | - Oronzo Brunetti
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (A.A.); (O.B.)
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95121 Catania, Italy;
| | - Nicola Silvestris
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (A.A.); (O.B.)
- Department of Biomedical Sciences and Human Oncology (DIMO), University of Bari, 70124 Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| |
Collapse
|
6
|
Giorello MB, Borzone FR, Labovsky V, Piccioni FV, Chasseing NA. Cancer-Associated Fibroblasts in the Breast Tumor Microenvironment. J Mammary Gland Biol Neoplasia 2021; 26:135-155. [PMID: 33398516 DOI: 10.1007/s10911-020-09475-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Years of investigation have shed light on a theory in which breast tumor epithelial cells are under the effect of the stromal microenvironment. This review aims to discuss recent findings concerning the phenotypic and functional characteristics of cancer associated fibroblasts (CAFs) and their involvement in tumor evolution, as well as their potential implications for anti-cancer therapy. In this manuscript, we reviewed that CAFs play a fundamental role in initiation, growth, invasion, and metastasis of breast cancer, and also serve as biomarkers in the clinical diagnosis, therapy, and prognosis of this disease.
Collapse
Affiliation(s)
- María Belén Giorello
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| | - Francisco Raúl Borzone
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Vivian Labovsky
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Flavia Valeria Piccioni
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos (IBYME) y Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Norma Alejandra Chasseing
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
7
|
Song T, Chen M, Wang X, Zhu E, Xue Y, Wang J, Sun B, Feng J. Intermittent hypoxia: Friend or foe on endothelial repair in mouse model. Exp Lung Res 2021; 47:211-225. [PMID: 33678107 DOI: 10.1080/01902148.2021.1891355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aim of the study: Obstructive sleep apnea, which is characterized by intermittent hypoxia (IH), is a common respiratory disease. The aim of the present study was to explore the relationship between hypoxia and endothelial progenitor cell (EPC) function, and explain the role of IH in endothelial repair.Materials and methods: Peripheral blood mononuclear cells (PBMCs) were isolated from a mouse model of IH. The number of CD133+ kinase insert domain receptor (KDR)+, CD133+CD34+, CD34+KDR+ and ALDHlowCD34+KDR+ EPCs was determined by flow cytometry. HIF-1α, stromal-derived factor-1 (SDF-1) α and VEGF were measured by ELISA. The proliferative ability of PBMCs was determined. EPC migration was assessed by Transwell assay and surface proteins by western blot analysis. EPCs were co-cultured with mouse brain endothelial cells and their angiogenic ability was analyzed.Results: The number of CD133+KDR+, CD133+CD34+ and CD34+KDR+ EPCs increased with IH ingravescence. The number of ALDHlowCD34+KDR+ EPCs with mild IH stimulation was higher and gradually decreased in the moderate and severe IH groups. The release of HIF-1α, SDF-1α and VEGF in the serum increased with the increase in the degree of IH. In the mild IH treatment, the migration and angiogenesis of EPCs, as well as the expression of vascular endothelial growth factor receptor 2 and cysteine-X-cysteine receptor 4, were higher than those in the control group, but progressively decreased in the groups with moderate and severe IH.Conclusion: Increased levels of IH accelerated the increase in vasoactive factors in peripheral blood, thereby mobilizing a large number of EPCs. Increasing of IH diminished the mobilization, chemotactic and angiogenetic ability of EPCs.
Collapse
Affiliation(s)
- Tao Song
- Intensive Care Unit of Tianjin Medical University General Hospital, Tianjin, China
| | - Mo Chen
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Endong Zhu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Yanchao Xue
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Juan Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Bei Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Jing Feng
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China.,Neuropharmacology Section, Laboratory of Toxicology & Pharmacology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| |
Collapse
|
8
|
Xie Y, Jiang Z, Yang R, Ye Y, Pei L, Xiong S, Wang S, Wang L, Liu S. Polysaccharide-rich extract from Polygonatum sibiricum protects hematopoiesis in bone marrow suppressed by triple negative breast cancer. Biomed Pharmacother 2021; 137:111338. [PMID: 33578234 DOI: 10.1016/j.biopha.2021.111338] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 01/19/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Polysaccharide is one of main components in Polygonatum sibiricum (PS), which is an herbal medicine widely used in East Asia. Polysaccharides from Polygonatum sibiricum has been shown to exhibit multiple biological activities, such as anti-diabetes, anti-inflammation, antioxidant, immunity modulation, and anticancer. Since hematopoietic system is one of determinant factors in cancer control, we here explored the effect of polysaccharide-rich extract from Polygonatum sibiricum (PREPS) on hematopoiesis in the mice bearing triple negative breast cancer (TNBC). We found that the 4T1 TNBC tumor significantly increased myeloid cells in peripheral blood, bone marrow and spleen, while decreasing bone marrow hematopoietic stem and progenitor cells (HSPCs), indicative of an inhibition of medullary hematopoiesis. When 4T1 TNBC tumor-bearing mice were treated with PREPS, the percentage of myeloid cells within tumor-infiltrating immune cells was reduced. In addition, PREPS also inhibited hematopoietic cell expansion in the spleen, which was induced by TNBC tumors. Importantly, PREPS markedly increased HSPCs and common lymphoid progenitors in the bone marrow that had been suppressed by TNBC tumors. These findings suggest that PREPS protect hematopoiesis inhibited by TNBC tumors in the bone marrow. Although PREPS alone did not achieve statistical significance in the suppression of TNBC tumor growth, it may have a long-lasting anti-tumor effect to assist TNBC therapies by sustaining hematopoiesis and lymphoid regeneration in bone marrow.
Collapse
Affiliation(s)
- Ying Xie
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Ziwei Jiang
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rui Yang
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiyi Ye
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lixia Pei
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Si Xiong
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shunchun Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lisheng Wang
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada.
| | - Sheng Liu
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
9
|
Halpern AL, Fitz JG, Fujiwara Y, Yi J, Anderson AL, Zhu Y, Schulick RD, El Kasmi KC, Barnett CC. Hepatic thermal injury promotes colorectal cancer engraftment in C57/black 6 mice. Am J Physiol Cell Physiol 2021; 320:C142-C151. [PMID: 33175574 PMCID: PMC7846977 DOI: 10.1152/ajpcell.00071.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 10/26/2020] [Accepted: 11/02/2020] [Indexed: 11/22/2022]
Abstract
Treatment options for liver metastases (primarily colorectal cancer) are limited by high recurrence rates and persistent tumor progression. Surgical approaches to management of these metastases typically use heat energy including electrocautery, argon beam coagulation, thermal ablation of surgical margins for hemostasis, and preemptive thermal ablation to prevent bleeding or to effect tumor destruction. Based on high rates of local recurrence, these studies assess whether local effects of hepatic thermal injury (HTI) might contribute to poor outcomes by promoting a hepatic microenvironment favorable for tumor engraftment or progression due to induction of procancer cytokines and deleterious immune infiltrates at the site of thermal injury. To test this hypothesis, an immunocompetent mouse model was developed wherein HTI was combined with concomitant intrasplenic injection of cells from a well-characterized MC38 colon carcinoma cell line. In this model, HTI resulted in a significant increase in engraftment and progression of MC38 tumors at the site of thermal injury. Furthermore, there were local increases in expression of messenger ribonucleic acid (mRNA) for hypoxia-inducible factor-1α (HIF1α), arginase-1, and vascular endothelial growth factor α and activation changes in recruited macrophages at the HTI site but not in untreated liver tissue. Inhibition of HIF1α following HTI significantly reduced discreet hepatic tumor development (P = 0.03). Taken together, these findings demonstrate that HTI creates a favorable local environment that is associated with protumorigenic activation of macrophages and implantation of circulating tumors. Discrete targeting of HIF1α signaling or inhibiting macrophages offers potential strategies for improving the outcome of surgical management of hepatic metastases where HTI is used.
Collapse
Affiliation(s)
- Alison L Halpern
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - J Gregory Fitz
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Yuki Fujiwara
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jeniann Yi
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Aimee L Anderson
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Yuwen Zhu
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Richard D Schulick
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Karim C El Kasmi
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Carlton C Barnett
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
10
|
Hurtado P, Martínez-Pena I, Piñeiro R. Dangerous Liaisons: Circulating Tumor Cells (CTCs) and Cancer-Associated Fibroblasts (CAFs). Cancers (Basel) 2020; 12:E2861. [PMID: 33027902 PMCID: PMC7599894 DOI: 10.3390/cancers12102861] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/02/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023] Open
Abstract
The crosstalk between cancer cells and the tumor microenvironment (TME) is a key determinant of cancer metastasis. Cancer-associated fibroblasts (CAFs), one of the main cellular components of TME, promote cancer cell invasion and dissemination through mechanisms including cell-cell interactions and the paracrine secretion of growth factors, cytokines and chemokines. During metastasis, circulating tumor cells (CTCs) are shed from the primary tumor to the bloodstream, where they can be detected as single cells or clusters. The current knowledge about the biology of CTC clusters positions them as key actors in metastasis formation. It also indicates that CTCs do not act alone and that they may be aided by stromal and immune cells, which seem to shape their metastatic potential. Among these cells, CAFs are found associated with CTCs in heterotypic CTC clusters, and their presence seems to increase their metastatic efficiency. In this review, we summarize the current knowledge on the role that CAFs play on metastasis and we discuss their implication on the biogenesis, metastasis-initiating capacity of CTC clusters, and clinical implications. Moreover, we speculate about possible therapeutic strategies aimed to limit the metastatic potential of CTC clusters involving the targeting of CAFs as well as their difficulties and limitations.
Collapse
Affiliation(s)
- Pablo Hurtado
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (P.H.); (I.M.-P.)
| | - Inés Martínez-Pena
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (P.H.); (I.M.-P.)
| | - Roberto Piñeiro
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (P.H.); (I.M.-P.)
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, 28029 Madrid, Spain
| |
Collapse
|
11
|
Leal V, Ribeiro CF, Oliveiros B, António N, Silva S. Intrinsic Vascular Repair by Endothelial Progenitor Cells in Acute Coronary Syndromes: an Update Overview. Stem Cell Rev Rep 2020; 15:35-47. [PMID: 30345477 DOI: 10.1007/s12015-018-9857-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Bone marrow-derived endothelial progenitor cells (EPCs) play a key role in the maintenance of endothelial homeostasis and endothelial repair at areas of vascular damage. The quantification of EPCs in peripheral blood by flow cytometry is a strategy to assess this reparative capacity. The number of circulating EPCs is inversely correlated with the number of cardiovascular risk factors and to the occurrence of cardiovascular events. Therefore, monitoring EPCs levels may provide an accurate assessment of susceptibility to cardiovascular injury, greatly improving risk stratification of patients with high cardiovascular risk, such as those with an acute myocardial infarction. However, there are many issues in the field of EPC identification and quantification that remain unsolved. In fact, there have been conflicting protocols used to the phenotypic identification of EPCs and there is still no consensual immunophenotypical profile that corresponds exactly to EPCs. In this paper we aim to give an overview on EPCs-mediated vascular repair with special focus on acute coronary syndromes and to discuss the different phenotypic profiles that have been used to identify and quantify circulating EPCs in several clinical studies. Finally, we will synthesize evidence on the prognostic role of EPCs in patients with high cardiovascular risk.
Collapse
Affiliation(s)
- Vânia Leal
- Group of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
| | - Carlos Fontes Ribeiro
- Institute of Pharmacology and Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Bárbara Oliveiros
- Laboratory of Biostatistics and Medical Informatics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Natália António
- Institute of Pharmacology and Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Cardiology Department, Coimbra Hospital and Universitary Centre, Coimbra, Portugal
| | - Sónia Silva
- Group of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.,Institute of Pharmacology and Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
12
|
Yuan J, Yuan B, Zeng L, Liu B, Chen Y, Meng X, Sun R, Lv X, Wang W, Yang S. Identification and validation of tumor microenvironment-related genes of prognostic value in lung adenocarcinoma. Oncol Lett 2020; 20:1772-1780. [PMID: 32724420 PMCID: PMC7377199 DOI: 10.3892/ol.2020.11735] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 03/17/2020] [Indexed: 12/30/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is a major subtype of non-small cell lung cancer. Despite significant progress in its diagnosis and treatment, the mortality and morbidity rate of LUAD remains high worldwide. The aim of the present study was to perform a systematic investigation of the tumor microenvironment (TME) and identify TME-related genes of prognostic value in patients with LUAD. Firstly, the immune scores and stromal scores of patients with LUAD from The Cancer Genome Atlas were calculated using the Estimation of STromal and Immune cells in MAlignant Tumors using Expression data algorithm, and a total of 281 prognostic TME-related genes were identified. Subsequently, functional analysis and protein-protein interaction network analysis revealed that these genes were mainly related to immune response, inflammatory response and chemotaxis. Finally, two independent LUAD cohorts from the Gene Expression Omnibus database were used to validate these genes, and 4 genes (GTPase IMAP family member 1, T-cell surface glycoprotein CD1b, integrin alpha-L and leukocyte surface antigen CD53) were identified, and downregulation of these genes was indicated to be associated with poor overall survival rate in patients with LUAD. In conclusion, a comprehensive analysis of TME was performed and 4 prognostic TME-related genes in patients with LUAD were identified.
Collapse
Affiliation(s)
- Jingyan Yuan
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Bo Yuan
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Lizhong Zeng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Boxuan Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yang Chen
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xia Meng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Ruiying Sun
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xin Lv
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Wei Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Shuanying Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
13
|
Hu P, Gao Y, Huang Y, Zhao Y, Yan H, Zhang J, Zhao L. Gene Expression-Based Immune Cell Infiltration Analyses of Prostate Cancer and Their Associations with Survival Outcome. DNA Cell Biol 2020; 39:1194-1204. [PMID: 32460527 DOI: 10.1089/dna.2020.5371] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer is the second most common cancer and the fifth cause of cancer death in males. Currently, there are no effective therapies for prostate cancer yet, and the status of treatment remains severe. In this study, we analyzed the composition of tumor-infiltrating immune cells (TIICs) in prostate cancer and paracancerous samples based on the gene expression profiles using CIBERSORT. Calculation of the TIIC subset proportions in 52 paired prostate cancer and paracancerous samples showed that their proportions were similar in intergroup and varied in intragroup. Compared with the paracancerous samples, the proportion of M0 macrophages was significantly increased in prostate cancer samples. Cox regression analysis using the TIIC subpopulations as continuous variables revealed that high plasma cell proportion was associated with poor 3-year Disease-Free Survival (DFS) in prostate cancer (hazard ratios = 1.8e-76, p = 0.001). Moreover, three immune clusters, which presented distinct prognosis, were identified using hierarchical clustering analysis based on the proportions of TIIC subpopulations. Among them, cluster 1 had superior 3-year DFS, while cluster 3 showed inferior 3-year DFS (p = 0.025). In summary, our research provided a comprehensive analysis on the TIIC composition in prostate cancer and suggested that both plasma cells and different cluster patterns were associated with the prostate cancer prognosis, which should be helpful for the clinical surveillance and treatment of prostate cancer.
Collapse
Affiliation(s)
- Ping Hu
- The Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Tianjin Key Laboratory of Cancer Prevention and Therapy; Tianjin's Clinical Research Center for Cancer, Tianjin, P.R. China.,The First Department of Medical Oncology, General Hospital of Ningxia Medical University, Yinchuan, P.R. China
| | - Yuanyuan Gao
- The Third Department of Medical Oncology, General Hospital of Ningxia Medical University, Yinchuan, P.R. China
| | - Ying Huang
- The Third Department of Medical Oncology, General Hospital of Ningxia Medical University, Yinchuan, P.R. China
| | - Yanjiao Zhao
- The Third Department of Medical Oncology, General Hospital of Ningxia Medical University, Yinchuan, P.R. China
| | - Hui Yan
- The Second Department of Medical Oncology, General Hospital of Ningxia Medical University, Yinchuan, P.R. China
| | - Jiao Zhang
- The First Department of Medical Oncology, General Hospital of Ningxia Medical University, Yinchuan, P.R. China
| | - Lujun Zhao
- The Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Tianjin Key Laboratory of Cancer Prevention and Therapy; Tianjin's Clinical Research Center for Cancer, Tianjin, P.R. China
| |
Collapse
|
14
|
Pluchino N, Mamillapalli R, Shaikh S, Habata S, Tal A, Gaye M, Taylor HS. CXCR4 or CXCR7 antagonists treat endometriosis by reducing bone marrow cell trafficking. J Cell Mol Med 2020; 24:2464-2474. [PMID: 31904910 PMCID: PMC7028867 DOI: 10.1111/jcmm.14933] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/29/2019] [Accepted: 11/25/2019] [Indexed: 12/18/2022] Open
Abstract
Adult stem cells have a major role in endometrial physiology, including remodelling and repair. However, they also have a critical role in the development and progression of endometriosis. Bone marrow-derived stem cells engraft eutopic endometrium and endometriotic lesions, differentiating to both stromal and epithelial cell fates. Using a mouse bone marrow transplantation model, we show that bone marrow-derived cells engrafting endometriosis express CXCR4 and CXCR7. Targeting either receptor by the administration of small molecule receptor antagonists AMD3100 or CCX771, respectively, reduced BM-derived stem cell recruitment into endometriosis implants. Endometriosis lesion size was decreased compared to vehicle controls after treatment with each antagonist in both an early growth and established lesion treatment model. Endometriosis lesion size was not effected when the local effects of CXCL12 were abrogated using uterine-specific CXCL12 null mice, suggesting an effect primarily on bone marrow cell migration rather than a direct endometrial effect. Antagonist treatment also decreased hallmarks of endometriosis physiopathology such as pro-inflammatory cytokine production and vascularization. CXCR4 and CXCR7 antagonists are potential novel, non-hormonal therapies for endometriosis.
Collapse
Affiliation(s)
- Nicola Pluchino
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Ramanaiah Mamillapalli
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Shafiq Shaikh
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Shutaro Habata
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Aya Tal
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Marie Gaye
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
15
|
Raskov H, Orhan A, Salanti A, Gögenur I. Premetastatic niches, exosomes and circulating tumor cells: Early mechanisms of tumor dissemination and the relation to surgery. Int J Cancer 2020; 146:3244-3255. [PMID: 31808150 DOI: 10.1002/ijc.32820] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/15/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022]
Abstract
The physiological stress response to surgery promotes wound healing and functional recovery and includes the activation of neural, inflammatory and proangiogenic signaling pathways. Paradoxically, the same pathways also promote metastatic spread and growth of residual cancer. Human and animal studies show that cancer surgery can increase survival, migration and proliferation of residual tumor cells. To secure the survival and growth of disseminated tumor cells, the formation of premetastatic niches in target organs involves a complex interplay between microenvironment, immune system, circulating tumor cells, as well as chemical mediators and exosomes secreted by the primary tumor. This review describes the current understanding of the early mechanisms of dissemination, as well as how surgery may facilitate disease progression.
Collapse
Affiliation(s)
- Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ali Salanti
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Wong AY, Whited JL. Parallels between wound healing, epimorphic regeneration and solid tumors. Development 2020; 147:147/1/dev181636. [PMID: 31898582 DOI: 10.1242/dev.181636] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Striking similarities between wound healing, epimorphic regeneration and the progression of solid tumors have been uncovered by recent studies. In this Review, we discuss systemic effects of tumorigenesis that are now being appreciated in epimorphic regeneration, including genetic, cellular and metabolic heterogeneity, changes in circulating factors, and the complex roles of immune cells and immune modulation at systemic and local levels. We suggest that certain mechanisms enabling regeneration may be co-opted by cancer to promote growth at primary and metastatic sites. Finally, we advocate that working with a unified approach could complement research in both fields.
Collapse
Affiliation(s)
- Alan Y Wong
- Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA 02138, USA
| | - Jessica L Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
17
|
Introduction - Biology of Breast Cancer Metastasis and Importance of the Analysis of CTCs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1220:1-10. [PMID: 32304076 DOI: 10.1007/978-3-030-35805-1_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Breast cancer metastasis is a complex multistep process during which tumor cells undergo structural and functional changes that allow them to move away from the primary tumor and disseminate to distant organs and tissues. Despite the inefficiency of this process, some populations of circulating tumor cells (CTCs), which are those cells responsible of metastases formation, are able to survive in blood circulation and grow into secondary tumors. Metastatic breast cancer remains an incurable disease, and the phenomenon of metastasis represents the larger cause of death in these patients. The application of liquid biopsy techniques and the advancements in the field have shown the prognostic value of CTCs, suggesting the importance that CTCs analyses may have in the clinic. However, their implementation in routine clinic has not been yet achieved due to the yet small body of evidence showing their clinical utility. This introductory chapter will revise the key aspects of breast cancer metastasis and discuss the importance of CTC analyses in the management of breast cancer patients.
Collapse
|
18
|
The Gastrointestinal Tumor Microenvironment: An Updated Biological and Clinical Perspective. JOURNAL OF ONCOLOGY 2019; 2019:6240505. [PMID: 31885581 PMCID: PMC6893275 DOI: 10.1155/2019/6240505] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/30/2019] [Indexed: 12/24/2022]
Abstract
Gastrointestinal cancers are still responsible for high numbers of cancer-related deaths despite advances in therapy. Tumor-associated cells play a key role in tumor biology, by supporting or halting tumor development through the production of extracellular matrix, growth factors, cytokines, and extracellular vesicles. Here, we review the roles of these tumor-associated cells in the initiation, angiogenesis, immune modulation, and resistance to therapy of gastrointestinal cancers. We also discuss novel diagnostic and therapeutic strategies directed at tumor-associated cells and their potential benefits for the survival of these patients.
Collapse
|
19
|
Alečković M, McAllister SS, Polyak K. Metastasis as a systemic disease: molecular insights and clinical implications. Biochim Biophys Acta Rev Cancer 2019; 1872:89-102. [PMID: 31202687 PMCID: PMC6692219 DOI: 10.1016/j.bbcan.2019.06.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/06/2019] [Accepted: 06/09/2019] [Indexed: 12/18/2022]
Abstract
Metastasis is a complex systemic disease that develops as a result of interactions between tumor cells and their local and distant microenvironments. Local and systemic immune-related changes play especially critical roles in limiting or enabling the development of metastatic disease. Although anti-tumor immune responses likely eliminate most early primary and metastatic lesions, factors secreted by cancer or stromal cells in the primary tumor can mobilize and activate cells in distant organs in a way that promotes the outgrowth of disseminated cancer cells into macrometastatic lesions. Therefore, the prevention, detection, and effective treatment of metastatic disease require a deeper understanding of the systemic effects of primary tumors as well as predisposing hereditary and acquired host factors including chronic inflammatory conditions. The success of immunotherapy in a subset of cancer patients is an example of how modulating the microenvironment and tumor-immune cell interactions can be exploited for the effective eradiation of even advanced-stage tumors. Here, we highlight emerging insights and clinical implications of cancer as a systemic disease.
Collapse
Affiliation(s)
- Maša Alečković
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America; Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Department of Medicine, Harvard Medical School, Boston, MA, United States of America
| | - Sandra S McAllister
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Department of Medicine, Harvard Medical School, Boston, MA, United States of America
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America; Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Department of Medicine, Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
20
|
Koustas E, Sarantis P, Kyriakopoulou G, Papavassiliou AG, Karamouzis MV. The Interplay of Autophagy and Tumor Microenvironment in Colorectal Cancer-Ways of Enhancing Immunotherapy Action. Cancers (Basel) 2019; 11:533. [PMID: 31013961 PMCID: PMC6520891 DOI: 10.3390/cancers11040533] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/07/2019] [Accepted: 04/11/2019] [Indexed: 02/05/2023] Open
Abstract
Autophagy as a primary homeostatic and catabolic process is responsible for the degradation and recycling of proteins and cellular components. The mechanism of autophagy has a crucial role in several cellular functions and its dysregulation is associated with tumorigenesis, tumor-stroma interactions, and resistance to cancer therapy. A growing body of evidence suggests that autophagy is also a key regulator of the tumor microenvironment and cellular immune response in different types of cancer, including colorectal cancer (CRC). Furthermore, autophagy is responsible for initiating the immune response especially when it precedes cell death. However, the role of autophagy in CRC and the tumor microenvironment remains controversial. In this review, we identify the role of autophagy in tumor microenvironment regulation and the specific mechanism by which autophagy is implicated in immune responses during CRC tumorigenesis and the context of anticancer therapy.
Collapse
Affiliation(s)
- Evangelos Koustas
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Georgia Kyriakopoulou
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Athanasios G Papavassiliou
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
- First Department of Internal Medicine, 'Laiko' General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| |
Collapse
|
21
|
Mizuno R, Kawada K, Itatani Y, Ogawa R, Kiyasu Y, Sakai Y. The Role of Tumor-Associated Neutrophils in Colorectal Cancer. Int J Mol Sci 2019; 20:ijms20030529. [PMID: 30691207 PMCID: PMC6386937 DOI: 10.3390/ijms20030529] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/24/2019] [Accepted: 01/24/2019] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common causes of cancer deaths worldwide and the number of CRC patients is increasing progressively. Despite the improvement of the surgical techniques and chemotherapy, we have not completely overcome this disease yet due to the metastases. Therefore, understanding the mechanisms through which metastasis occurs is important for overcoming CRC. Normal host cells in the tumor microenvironment, such as macrophages and fibroblasts, have been reported to promote the growth of CRCs. Although neutrophils were originally considered to have defensive functions against tumor cells, it has been revealed that some populations of neutrophils, called as tumor-associated neutrophils (TANs), have tumor-supportive functions. The plasticity between tumor-suppressive and -supportive neutrophils are regulated by transforming growth factor (TGF)-β and Interferon-β signaling. Some studies have demonstrated that TANs promote the spread of cancer cells to distant organs. TANs contribute to the tumor invasion and angiogenesis through the production of matrix metalloproteinase-9 (MMP9), vascular endothelial growth factor (VEGF), and hepatocyte growth factor (HGF) in the primary and metastatic sites. Neutrophils also promotes tumor cell dissemination by capturing circulating tumor cells using neutrophil extracellular traps and promote their migration to distant sites. The neutrophil-to-lymphocyte ratio is a well-defined predictive marker for CRC patients. In this review, we highlight the molecular signaling between TANs and CRC cells and the possibility of TANs as a potential target for cancer therapy.
Collapse
Affiliation(s)
- Rei Mizuno
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.
| | - Kenji Kawada
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.
| | - Yoshiro Itatani
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.
| | - Ryotaro Ogawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.
| | - Yoshiyuki Kiyasu
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.
| | - Yoshiharu Sakai
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
22
|
Adjei IM, Temples MN, Brown SB, Sharma B. Targeted Nanomedicine to Treat Bone Metastasis. Pharmaceutics 2018; 10:E205. [PMID: 30366428 PMCID: PMC6320768 DOI: 10.3390/pharmaceutics10040205] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/15/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023] Open
Abstract
Bone metastases are common complications of solid tumors, particularly those of the prostate, breast, and lungs. Bone metastases can lead to painful and devastating skeletal-related events (SREs), such as pathological fractures and nerve compressions. Despite advances in treatment for cancers in general, options for bone metastases remain inadequate and generally palliative. Anticancer drugs (chemotherapy and radiopharmaceuticals) do not achieve therapeutic concentrations in the bone and are associated with dose-limiting side effects to healthy tissues. Nanomedicines, with their tunable characteristics, have the potential to improve drug targeting to bone metastases while decreasing side effects for their effective treatment. In this review, we present the current state of the art for nanomedicines to treat bone metastases. We also discuss new treatment modalities enhanced by nanomedicine and their effects on SREs and disease progression.
Collapse
Affiliation(s)
- Isaac M Adjei
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville 32611, FL, USA.
| | - Madison N Temples
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville 32611, FL, USA.
| | - Shannon B Brown
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville 32611, FL, USA.
| | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville 32611, FL, USA.
| |
Collapse
|
23
|
ADAMTS1 protease is required for a balanced immune cell repertoire and tumour inflammatory response. Sci Rep 2018; 8:13103. [PMID: 30166561 PMCID: PMC6117274 DOI: 10.1038/s41598-018-31288-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022] Open
Abstract
Recent advances have emphasized the relevance of studying the extracellular microenvironment given its main contribution to tissue homeostasis and disease. Within this complex scenario, we have studied the extracellular protease ADAMTS1 (a disintegrin and metalloprotease with thrombospondin motif 1), implicated in vascularization and development, with reported anti- and pro-tumorigenic activities. In this work we performed a detailed study of the vasculature and substrates in adult organs of wild type and Adamts1-deficient mice. In addition to the expected alterations of organs like kidney, heart and aorta, we found that the lack of ADAMTS1 differently affects lymphocyte and myeloid populations in the spleen and bone marrow. The study of the substrate versican also revealed its alteration in the absence of the protease. With such premises, we challenged our mice with subcutaneous B16F1 syngeneic tumours and closely evaluated the immune repertoire in the tumours but also in the distant spleen and bone marrow. Our results confirmed a pro-inflammatory landscape in the absence of ADAMTS1, correlating with tumour blockade, supporting its novel role as a modulator of the immune cell response.
Collapse
|
24
|
Ubellacker JM, Baryawno N, Severe N, DeCristo MJ, Sceneay J, Hutchinson JN, Haider MT, Rhee CS, Qin Y, Gregory WM, Garrido-Castro AC, Holen I, Brown JE, Coleman RE, Scadden DT, McAllister SS. Modulating Bone Marrow Hematopoietic Lineage Potential to Prevent Bone Metastasis in Breast Cancer. Cancer Res 2018; 78:5300-5314. [PMID: 30065048 DOI: 10.1158/0008-5472.can-18-0548] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 06/12/2018] [Accepted: 07/23/2018] [Indexed: 12/20/2022]
Abstract
The presence of disseminated tumor cells in breast cancer patient bone marrow aspirates predicts decreased recurrence-free survival. Although it is appreciated that physiologic, pathologic, and therapeutic conditions impact hematopoiesis, it remains unclear whether targeting hematopoiesis presents opportunities for limiting bone metastasis. Using preclinical breast cancer models, we discovered that marrow from mice treated with the bisphosphonate zoledronic acid (ZA) are metastasis-suppressive. Specifically, ZA modulated hematopoietic myeloid/osteoclast progenitor cell (M/OCP) lineage potential to activate metastasis-suppressive activity. Granulocyte-colony stimulating factor (G-CSF) promoted ZA resistance by redirecting M/OCP differentiation. We identified M/OCP and bone marrow transcriptional programs associated with metastasis suppression and ZA resistance. Analysis of patient blood samples taken at randomization revealed that women with high-plasma G-CSF experienced significantly worse outcome with adjuvant ZA than those with lower G-CSF levels. Our findings support discovery of therapeutic strategies to direct M/OCP lineage potential and biomarkers that stratify responses in patients at risk of recurrence.Significance: Bone marrow myeloid/osteoclast progenitor cell lineage potential has a profound impact on breast cancer bone metastasis and can be modulated by G-CSF and bone-targeting agents. Cancer Res; 78(18); 5300-14. ©2018 AACR.
Collapse
Affiliation(s)
- Jessalyn M Ubellacker
- Hematology Division, Brigham & Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Ninib Baryawno
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.,Center for Regenerative Medicine and the Cancer Center, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Nicolas Severe
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.,Center for Regenerative Medicine and the Cancer Center, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Molly J DeCristo
- Hematology Division, Brigham & Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Jaclyn Sceneay
- Hematology Division, Brigham & Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - John N Hutchinson
- Department of Biostatistics, Harvard T.H. Chan, School of Public Health, Boston, Massachusetts
| | - Marie-Therese Haider
- Academic Unit of Clinical Oncology, Department of Oncology & Metabolism, Weston Park Hospital, University of Sheffield, Sheffield, United Kingdom
| | - Catherine S Rhee
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.,Center for Regenerative Medicine and the Cancer Center, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Yuanbo Qin
- Hematology Division, Brigham & Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Walter M Gregory
- Clinical Trials Research Unit, University of Leeds, Leeds, United Kingdom
| | - Ana C Garrido-Castro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ingunn Holen
- Academic Unit of Clinical Oncology, Department of Oncology & Metabolism, Weston Park Hospital, University of Sheffield, Sheffield, United Kingdom
| | - Janet E Brown
- Academic Unit of Clinical Oncology, Department of Oncology & Metabolism, Weston Park Hospital, University of Sheffield, Sheffield, United Kingdom
| | - Robert E Coleman
- Academic Unit of Clinical Oncology, Department of Oncology & Metabolism, Weston Park Hospital, University of Sheffield, Sheffield, United Kingdom
| | - David T Scadden
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.,Center for Regenerative Medicine and the Cancer Center, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Sandra S McAllister
- Hematology Division, Brigham & Women's Hospital, Boston, Massachusetts. .,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| |
Collapse
|
25
|
Wang G, Zhang L, Zhou Y, Sun Q, Xu H, Cai F, Xiang P, Chen Z, Jiang H. KAI1/CD82 Genetically Engineered Endothelial Progenitor Cells Inhibit Metastasis of Human Nasopharyngeal Carcinoma in a Mouse Model. Med Sci Monit 2018; 24:3146-3152. [PMID: 29755107 PMCID: PMC5975073 DOI: 10.12659/msm.907219] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Endothelial progenitor cells (EPCs) are regarded as promising targeted vectors for delivering therapeutic genes or agents in cancer therapy. The purpose of this study was to investigate the role of intravenously administered KAI1/CD82 genetically transduced EPCs in the tumorigenesis and metastasis of nasopharyngeal carcinoma (NPC). Material/Methods EPCs were isolated from human umbilical cord blood, expanded in culture, and stably transduced with lentiviral vectors expressing KAI1/CD82. The KAI1/CD82 EPCs were injected intravenously into nude mice bearing human NPC xenografts. Tumor growth and the incidence of liver and lung metastases were observed. Expression of KAI1/CD82 was determined by immunofluorescent staining. Results The NPC model was successfully established. Tumor growth was not suppressed when mice were injected with KAI1/CD82 EPCs (KAI1/CD82 EPCs group) compared with when non-transduced EPCs was present (EPCs group) or the control (1.485±0.234, 1.388±0.204, and 1.487±0.223g, respectively; P>0.05). However, the incidence of lung metastasis was significantly reduced in the KAI1/CD82+ EPCs group compared with the EPCs group and the control group (10%, 55% and 45%, respectively; P=0.005), and there was a significant decrease in the number of metastatic foci on the lung surface (17.50±3.54, 34.27±5.35, and 38.44±9.63 respectively; P=0.007). Moreover, KAI1/CD82 was expressed in lung metastatic foci of the KAI1/CD82 EPCs group, but not in the EPCs group and control group. Conclusions EPCs can be used as a delivery vehicle for suppressor genes KAI1/CD82 to NPC, and the migration of KAI1/CD82 genetically engineered EPCs can inhibit NPC lung metastasis in a mouse model.
Collapse
Affiliation(s)
- Gengming Wang
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China (mainland)
| | - Lei Zhang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China (mainland)
| | - Yan Zhou
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China (mainland)
| | - Qian Sun
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China (mainland)
| | - Hongbo Xu
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China (mainland)
| | - Feng Cai
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China (mainland)
| | - Ping Xiang
- Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China (mainland)
| | - Zhendong Chen
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Bengbu, Anhui, China (mainland)
| | - Hao Jiang
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China (mainland)
| |
Collapse
|
26
|
Redundant angiogenic signaling and tumor drug resistance. Drug Resist Updat 2018; 36:47-76. [DOI: 10.1016/j.drup.2018.01.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/22/2017] [Accepted: 01/11/2018] [Indexed: 02/07/2023]
|
27
|
Wu Y, Gao B, Xiong QJ, Wang YC, Huang DK, Wu WN. Acid-sensing ion channels contribute to the effect of extracellular acidosis on proliferation and migration of A549 cells. Tumour Biol 2017; 39:1010428317705750. [PMID: 28618956 DOI: 10.1177/1010428317705750] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Acid-sensing ion channels, a proton-gated cation channel, can be activated by low extracellular pH and involved in pathogenesis of some tumors such as glioma and breast cancer. However, the role of acid-sensing ion channels in the growth of lung cancer cell is unclear. In this study, we investigated the expression of acid-sensing ion channels in human lung cancer cell line A549 and their possible role in proliferation and migration of A549 cells. The results show that acid-sensing ion channel 1, acid-sensing ion channel 2, and acid-sensing ion channel 3 are expressed in A549 cells at the messenger RNA and protein levels, and acid-sensing ion channel-like currents were elicited by extracellular acid stimuli. Moreover, we found that acidic extracellular medium or overexpressing acid-sensing ion channel 1a promotes proliferation and migration of A549 cells. In addition psalmotoxin 1, a specific acid-sensing ion channel 1a inhibitor, or acid-sensing ion channel 1a knockdown can abolish the effect of acid stimuli on A549 cells. In addition, acid-sensing ion channels mediate increase of [Ca2+]i induced by low extracellular pH in A549 cells. All these results indicate that acid-sensing ion channel-calcium signal mediate lung cancer cell proliferation and migration induced by extracellular acidosis, and acid-sensing ion channels may serve as a prognostic marker and a therapeutic target for lung cancer.
Collapse
Affiliation(s)
- Yu Wu
- 1 Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, P.R. China
| | - Bo Gao
- 1 Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, P.R. China
| | - Qiu-Ju Xiong
- 2 Department of Pain Management, Wuhan Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yu-Chan Wang
- 1 Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, P.R. China
| | - Da-Ke Huang
- 3 Synthetic Laboratory, School of Basic Medical Sciences, Anhui Medical University, Hefei, P.R. China
| | - Wen-Ning Wu
- 1 Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, P.R. China.,4 Key Laboratory of Anti-inflammatory and Immunopharmacology, Anhui Medical University, Hefei, P.R. China
| |
Collapse
|
28
|
Khatun M, Sorjamaa A, Kangasniemi M, Sutinen M, Salo T, Liakka A, Lehenkari P, Tapanainen JS, Vuolteenaho O, Chen JC, Lehtonen S, Piltonen TT. Niche matters: The comparison between bone marrow stem cells and endometrial stem cells and stromal fibroblasts reveal distinct migration and cytokine profiles in response to inflammatory stimulus. PLoS One 2017; 12:e0175986. [PMID: 28419140 PMCID: PMC5395216 DOI: 10.1371/journal.pone.0175986] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/03/2017] [Indexed: 12/17/2022] Open
Abstract
Objective Intrinsic inflammatory characteristics play a pivotal role in stem cell recruitment and homing through migration where the subsequent change in niche has been shown to alter these characteristics. The bone marrow mesenchymal stem cells (bmMSCs) have been demonstrated to migrate to the endometrium contributing to the stem cell reservoir and regeneration of endometrial tissue. Thus, the aim of the present study was to compare the inflammation-driven migration and cytokine secretion profile of human bmMSCs to endometrial mesenchymal stem cells (eMSCs) and endometrial fibroblasts (eSFs). Materials and methods The bmMSCs were isolated from bone marrow aspirates through culturing, whereas eMSCs and eSFs were FACS-isolated. All cell types were tested for their surface marker, proliferation profiles and migration properties towards serum and inflammatory attractants. The cytokine/chemokine secretion profile of 35 targets was analysed in each cell type at basal level along with lipopolysaccharide (LPS)-induced state. Results Both stem cell types, bmMSCs and eMSCs, presented with similar stem cell surface marker profiles as well as possessed high proliferation and migration potential compared to eSFs. In multiplex assays, the secretion of 16 cytokine targets was detected and LPS stimulation expanded the cytokine secretion pattern by triggering the secretion of several targets. The bmMSCs exhibited higher cytokine secretion of vascular endothelial growth factor (VEGF)-A, stromal cell-derived factor-1 alpha (SDF)-1α, interleukin-1 receptor antagonist (IL-1RA), IL-6, interferon-gamma inducible protein (IP)-10, monocyte chemoattractant protein (MCP)-1, macrophage inflammatory protein (MIP)1α and RANTES compared to eMSCs and/or eSFs after stimulation with LPS. The basal IL-8 secretion was higher in both endometrial cell types compared to bmMSCs. Conclusion Our results highlight that similar to bmMSCs, the eMSCs possess high migration activity while the differentiation process towards stromal fibroblasts seemed to result in loss of stem cell surface markers, minimal migration activity and a subtler cytokine profile likely contributing to normal endometrial function.
Collapse
Affiliation(s)
- Masuma Khatun
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Anna Sorjamaa
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Marika Kangasniemi
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Meeri Sutinen
- Cancer and Translational Medicine Research Unit, Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Tuula Salo
- Cancer and Translational Medicine Research Unit, Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Annikki Liakka
- Department of Pathology, Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Petri Lehenkari
- Department of Anatomy and Department of Internal Medicine, Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Juha S. Tapanainen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | | | - Joseph C. Chen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, United States of America
| | - Siri Lehtonen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Terhi T. Piltonen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland
- * E-mail:
| |
Collapse
|
29
|
Dai J, Lu Y, Roca H, Keller JM, Zhang J, McCauley LK, Keller ET. Immune mediators in the tumor microenvironment of prostate cancer. CHINESE JOURNAL OF CANCER 2017; 36:29. [PMID: 28292326 PMCID: PMC5351274 DOI: 10.1186/s40880-017-0198-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 03/09/2017] [Indexed: 01/24/2023]
Abstract
Prostate cancer tissue is composed of both cancer cells and host cells. The milieu of host components that compose the tumor is termed the tumor microenvironment (TME). Host cells can be those derived from the tissue in which the tumor originates (e.g., fibroblasts and endothelial cells) or those recruited, through chemotactic or other factors, to the tumor (e.g., circulating immune cells). Some immune cells are key players in the TME and represent a large proportion of non-tumor cells found within the tumor. Immune cells can have both anti-tumor and pro-tumor activity. In addition, crosstalk between prostate cancer cells and immune cells affects immune cell functions. In this review, we focus on immune cells and cytokines that contribute to tumor progression. We discuss T-regulatory and T helper 17 cells and macrophages as key modulators in prostate cancer progression. In addition, we discuss the roles of interleukin-6 and receptor activator of nuclear factor kappa-B ligand in modulating prostate cancer progression. This review highlights the concept that immune cells and cytokines offer a potentially promising target for prostate cancer therapy.
Collapse
Affiliation(s)
- Jinlu Dai
- Department of Urology and Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yi Lu
- Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi, 520021, P. R. China
| | - Hernan Roca
- Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jill M Keller
- Department of Urology and Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jian Zhang
- Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi, 520021, P. R. China
| | - Laurie K McCauley
- Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Evan T Keller
- Department of Urology and Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA. .,Department of Urology, University of Michigan, Ann Arbor, MI, 48109-8940, USA.
| |
Collapse
|
30
|
Ubellacker JM, Haider MT, DeCristo MJ, Allocca G, Brown NJ, Silver DP, Holen I, McAllister SS. Zoledronic acid alters hematopoiesis and generates breast tumor-suppressive bone marrow cells. Breast Cancer Res 2017; 19:23. [PMID: 28264701 PMCID: PMC5339994 DOI: 10.1186/s13058-017-0815-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 02/09/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The bone-targeting agent zoledronic acid (ZOL) increases breast cancer survival in subsets of patients, but the underlying reasons for this protective effect are unknown. ZOL modulates the activity of osteoclasts and osteoblasts, which form hematopoietic stem cell niches, and therefore may affect hematopoietic cells that play a role in breast cancer progression. METHOD Immunocompetent and immunocompromised strains of mice commonly used for breast cancer research were injected with a single, clinically relevant dose of ZOL (100 μg/kg) or vehicle control. The effects of ZOL on the bone marrow microenvironment (bone volume, bone cell number/activity, extracellular matrix composition) were established at various time points following treatment, using micro-computed tomography (μCT) analysis, histomorphometry, ELISA and immunofluorescence. The effects on peripheral blood and bone marrow hematopoietic progenitor populations were assessed using a HEMAVET® hematology analyzer and multicolor flow cytometry, respectively. Tumor support function of bone marrow cells was determined using an in vivo functional assay developed in our laboratory. RESULTS Using multiple mouse strains, we observed transient changes in numbers of hematopoietic stem cells, myeloid-biased progenitor cells, and lymphoid-biased cells concurrent with changes to hematopoietic stem cell niches following ZOL administration. Importantly, bone marrow cells from mice treated with a single, clinically relevant dose of ZOL inhibited breast tumor outgrowth in vivo. The ZOL-induced tumor suppressive function of the bone marrow persisted beyond the time point at which numbers of hematopoietic progenitor cells had returned to baseline. CONCLUSIONS These findings provide novel evidence that alterations to the bone marrow play a role in the anti-tumor activity of ZOL and suggest possibilities for capitalizing on the beneficial effects of ZOL in reducing breast cancer development and progression.
Collapse
Affiliation(s)
- Jessalyn M. Ubellacker
- Department of Medicine, Harvard Medical School, Boston, MA 02115 USA
- Hematology Division, Brigham & Women’s Hospital, Boston, MA 02115 USA
| | | | - Molly J. DeCristo
- Department of Medicine, Harvard Medical School, Boston, MA 02115 USA
- Hematology Division, Brigham & Women’s Hospital, Boston, MA 02115 USA
| | - Gloria Allocca
- Department of Oncology & Metabolism, University of Sheffield, Sheffield, UK
| | - Nicola J. Brown
- Department of Oncology & Metabolism, University of Sheffield, Sheffield, UK
| | - Daniel P. Silver
- Departments of Medical Oncology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Ingunn Holen
- Department of Oncology & Metabolism, University of Sheffield, Sheffield, UK
| | - Sandra S. McAllister
- Department of Medicine, Harvard Medical School, Boston, MA 02115 USA
- Hematology Division, Brigham & Women’s Hospital, Boston, MA 02115 USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA
- Harvard Stem Cell Institute, Cambridge, MA 02138 USA
| |
Collapse
|
31
|
Abstract
Adult stem cells have a major role in endometrial physiology, remodeling, and repair, but they also have a critical role in the development and progression of endometriosis. Bone marrow-derived stem cells (BMDSCs) engraft eutopic endometrium and endometriotic lesions, showing stromal and epithelial fate. Nevertheless, circulating BMDSCs are in limited supply, and the presence of endometriosis depletes stem cells from the blood circulation, preventing their homing in the uterus. Furthermore, stem cells migrate from endometriotic lesion into the uterus, leading to a dysfunctional endometrium. Stem cell trafficking is a central feature of endometriosis. Understanding molecular mechanisms regulating cell mobility and engraftment in endometriosis may reveal new targets for treatment.
Collapse
Affiliation(s)
- Nicola Pluchino
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
32
|
Yang X, Hao J, Mao Y, Jin ZQ, Cao R, Zhu CH, Liu XH, Liu C, Ding XL, Wang XD, Chen D, Wu XZ. bFGF Promotes Migration and Induces Cancer-Associated Fibroblast Differentiation of Mouse Bone Mesenchymal Stem Cells to Promote Tumor Growth. Stem Cells Dev 2016; 25:1629-1639. [PMID: 27484709 DOI: 10.1089/scd.2016.0217] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Tumors recruit bone mesenchymal stem cells (BMSCs) to localize to tumor sites, which induces their conversion into cancer-associated fibroblasts (CAFs) that facilitate tumor progression. However, this process is poorly understood on the molecular level. In this study, we found that 4T1 breast cancer cells promoted the migration of BMSCs, and bFGF neutralizing antibody inhibited the migration of BMSCs induced by a tumor-conditioned medium. In addition, exogenous bFGF enhanced the migration of BMSCs in a dose-dependent manner in vitro. Furthermore, BMSCs promoted the proliferation of 4T1 tumor cells under BMSC-conditioned medium and in tumor xenograft model. Dramatically, BMSCs expressed CAF markers and produced collagen in the tumor microenvironment, and this transition was blocked by bFGF antibody. In addition, exogenous bFGF induced CAF differentiation of BMSCs. And bFGF increased phosphorylation of Erk1/2 and Smad3 in BMSCs and Erk inhibitor PD98059 was shown to block bFGF-induced Erk and Smad3 phosphorylation, suggesting that Erk/Smad3 signaling pathway involved in BMSC transdifferentiation induced by bFGF. Collectively, our results indicate that bFGF signaling plays indispensable roles in BMSC recruitment and transdifferentiation into CAFs and the consequent protumor effects, and targeting tumor stroma through bFGF inhibition maybe a promising strategy to suppress tumor progression.
Collapse
Affiliation(s)
- Xue Yang
- 1 Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital , Tianjin, China
| | - Jian Hao
- 1 Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital , Tianjin, China
| | - Yu Mao
- 1 Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital , Tianjin, China
| | - Zi-Qi Jin
- 2 Tianjin Medical University , Tianjin, China
| | - Rui Cao
- 1 Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital , Tianjin, China
| | - Cui-Hong Zhu
- 3 Zhong-Shan-Men In-Patient Department, Tianjin Medical University Cancer Institute and Hospital , Tianjin, China
| | - Xiao-Hui Liu
- 1 Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital , Tianjin, China
| | - Chang Liu
- 3 Zhong-Shan-Men In-Patient Department, Tianjin Medical University Cancer Institute and Hospital , Tianjin, China
| | - Xiu-Li Ding
- 1 Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital , Tianjin, China
| | - Xiao-Dong Wang
- 4 Tianjin Medical University General Hospital , Tianjin, China
| | - Dan Chen
- 2 Tianjin Medical University , Tianjin, China
| | - Xiong-Zhi Wu
- 1 Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital , Tianjin, China
| |
Collapse
|
33
|
Bone marrow-derived cells are recruited by the melanoma tumor with endothelial cells contributing to tumor vasculature. Clin Transl Oncol 2016; 19:125-133. [PMID: 27188167 DOI: 10.1007/s12094-016-1515-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 04/19/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE Tumor expansion is dependent on neovascularization, a process that requires sustained new vessel formation. Although the critical role of angiogenesis by endothelial sprouting in this process, controversy still prevails on whether angiogenesis involving bone marrow-derived endothelial cells, does contribute to this process. This study aims to evaluate the recruitment of bone marrow-derived cells by the melanoma tumor, including endothelial cells, and if they contribute to angiogenesis. METHODS A chimeric mouse model of GFP bone marrow was used to induce melanoma tumors derived from murine B16-F10 cell line. These tumors were evaluated for the presence of myeloid cells (CD11b), T lymphocytes (CD3, CD4 and CD8) and endothelial cells (VEGFR2 and CD31) derived from bone marrow. RESULTS Mice transplanted with GFP+ cells showed significant bone marrow chimerism (90.9 ± 0.87 %) when compared to the GFP transgenic mice (90.66 ± 2.1 %, p = 0.83) demonstrating successful engraftment of donor bone marrow stem/progenitor cells. Analysis of the murine melanoma tumor showed the presence of donor cells in the tumors (3.5 ± 1.7 %) and interestingly, these cells represent endothelial cells (CD31+ cells; 11.5 ± 6.85 %) and myeloid cells (CD11b+ cells; 80 ± 21 %), but also tumor-infiltrating lymphocytes (CD8+ T cells, 13.31 ± 0.2 %; CD4+ T-cells, 2.1 ± 1.2 %). Examination of the tumor endothelium by confocal microscopy suggests the presence of donor CD31+/GFP+ cells in the wall of some blood vessels. CONCLUSION This study demonstrates that bone marrow-derived cells are recruited by the murine melanoma tumor, with myeloid cells and CD4 and CD8 T lymphocytes migrating as antitumor immune response, and endothelial cells participating of the tumor blood vessels formation.
Collapse
|
34
|
Ubellacker JM, McAllister SS. The unresolved role of systemic factors in bone metastasis. J Bone Oncol 2016; 5:96-99. [PMID: 27761365 PMCID: PMC5063227 DOI: 10.1016/j.jbo.2016.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/10/2016] [Accepted: 03/14/2016] [Indexed: 12/19/2022] Open
Abstract
Systemic factors including cytokines, cell-free nucleic acids, microvesicles, and platelets are appreciated as important regulators of adenocarcinoma progression. Research findings using pre-clinical mouse models have revealed that many such systemically acting factors are either secreted by or responsive to peripheral tumors and impact bone and bone marrow (collectively referred to as the bone microenvironment) to initiate processes that ultimately govern disease progression, even in the absence of detectable bone metastases. In some cases, cancer-driven modulation of the bone microenvironment involves mobilization of bone marrow hematopoietic and mesenchymal cells into the circulation that are subsequently recruited into peripheral tissues and tumors. In other cases, systemic factors alter bone marrow cell (BMC) differentiation and/or gene expression to render the BMCs pro-tumorigenic even prior to their mobilization into the circulation. Given their effect on the bone microenvironment, it stands to reason that such systemic factors might also influence metastases in the bone; however, this hypothesis remains to be comprehensively tested. Here, we briefly review what is known, and not known, about systemic factors that regulate the bone microenvironment and thereby influence bone metastases. We also pose a number of currently unanswered questions in this active area of research. A better understanding of systemic processes that influence bone metastasis should aid discovery of therapeutic approaches that aim to eradicate or reduce disease burden in the bone, which is the cause of significant patient mortality and morbidity and is currently incurable.
Collapse
Affiliation(s)
| | - Sandra S McAllister
- Harvard Medical School, Boston, MA 02115, USA; Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
35
|
The Role of Chemokines in Promoting Colorectal Cancer Invasion/Metastasis. Int J Mol Sci 2016; 17:ijms17050643. [PMID: 27136535 PMCID: PMC4881469 DOI: 10.3390/ijms17050643] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 04/18/2016] [Accepted: 04/25/2016] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related death worldwide. Although most of the primary CRC can be removed by surgical resection, advanced tumors sometimes show recurrences in distant organs such as the liver, lung, lymph node, bone or peritoneum even after complete resection of the primary tumors. In these advanced and metastatic CRC, it is the tumor-stroma interaction in the tumor microenvironment that often promotes cancer invasion and/or metastasis through chemokine signaling. The tumor microenvironment contains numerous host cells that may suppress or promote cancer aggressiveness. Several types of host-derived myeloid cells reside in the tumor microenvironment, and the recruitment of them is under the control of chemokine signaling. In this review, we focus on the functions of chemokine signaling that may affect tumor immunity by recruiting several types of bone marrow-derived cells (BMDC) to the tumor microenvironment of CRC.
Collapse
|
36
|
Marsh T, Wong I, Sceneay J, Barakat A, Qin Y, Sjödin A, Alspach E, Nilsson B, Stewart SA, McAllister SS. Hematopoietic Age at Onset of Triple-Negative Breast Cancer Dictates Disease Aggressiveness and Progression. Cancer Res 2016; 76:2932-43. [PMID: 27197230 DOI: 10.1158/0008-5472.can-15-3332] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/18/2016] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) is considered an early onset subtype of breast cancer that carries with it a poorer prognosis in young rather than older women for reasons that remain poorly understood. Hematopoiesis in the bone marrow becomes altered with age and may therefore affect the composition of tumor-infiltrating hematopoietic cells and subsequent tumor progression. In this study, we investigated how age- and tumor-dependent changes to bone marrow-derived hematopoietic cells impact TNBC progression. Using multiple mouse models of TNBC tumorigenesis and metastasis, we found that a specific population of bone marrow cells (BMC) upregulated CSF-1R and secreted the growth factor granulin to support stromal activation and robust tumor growth in young mice. However, the same cell population in old mice expressed low levels of CSF1R and granulin and failed to promote tumor outgrowth, suggesting that age influences the tumorigenic capacity of BMCs in response to tumor-associated signals. Importantly, BMCs from young mice were sufficient to activate a tumor-supportive microenvironment and induce tumor progression in old mice. These results indicate that hematopoietic age is an important determinant of TNBC aggressiveness and provide rationale for investigating age-stratified therapies designed to prevent the protumorigenic effects of activated BMCs. Cancer Res; 76(10); 2932-43. ©2016 AACR.
Collapse
Affiliation(s)
- Timothy Marsh
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Irene Wong
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Jaclyn Sceneay
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Amey Barakat
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Yuanbo Qin
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Andreas Sjödin
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts
| | - Elise Alspach
- Department of Cell Biology and Physiology; Department of Medicine; and ICCE Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Björn Nilsson
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden. Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Sheila A Stewart
- Department of Cell Biology and Physiology; Department of Medicine; and ICCE Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Sandra S McAllister
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts. Broad Institute of Harvard and MIT, Cambridge, Massachusetts. Harvard Stem Cell Institute, Cambridge, Massachusetts.
| |
Collapse
|
37
|
Mittal V, El Rayes T, Narula N, McGraw TE, Altorki NK, Barcellos-Hoff MH. The Microenvironment of Lung Cancer and Therapeutic Implications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 890:75-110. [PMID: 26703800 DOI: 10.1007/978-3-319-24932-2_5] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The tumor microenvironment (TME) represents a milieu that enables tumor cells to acquire the hallmarks of cancer. The TME is heterogeneous in composition and consists of cellular components, growth factors, proteases, and extracellular matrix. Concerted interactions between genetically altered tumor cells and genetically stable intratumoral stromal cells result in an "activated/reprogramed" stroma that promotes carcinogenesis by contributing to inflammation, immune suppression, therapeutic resistance, and generating premetastatic niches that support the initiation and establishment of distant metastasis. The lungs present a unique milieu in which tumors progress in collusion with the TME, as evidenced by regions of aberrant angiogenesis, acidosis and hypoxia. Inflammation plays an important role in the pathogenesis of lung cancer, and pulmonary disorders in lung cancer patients such as chronic obstructive pulmonary disease (COPD) and emphysema, constitute comorbid conditions and are independent risk factors for lung cancer. The TME also contributes to immune suppression, induces epithelial-to-mesenchymal transition (EMT) and diminishes efficacy of chemotherapies. Thus, the TME has begun to emerge as the "Achilles heel" of the disease, and constitutes an attractive target for anti-cancer therapy. Drugs targeting the components of the TME are making their way into clinical trials. Here, we will focus on recent advances and emerging concepts regarding the intriguing role of the TME in lung cancer progression, and discuss future directions in the context of novel diagnostic and therapeutic opportunities.
Collapse
MESH Headings
- Antibodies, Monoclonal/therapeutic use
- Antineoplastic Agents/therapeutic use
- Carcinogenesis/drug effects
- Carcinogenesis/genetics
- Carcinogenesis/metabolism
- Carcinogenesis/pathology
- Cell Communication/drug effects
- Drug Resistance, Neoplasm/genetics
- Epithelial-Mesenchymal Transition/drug effects
- Epithelial-Mesenchymal Transition/genetics
- Gene Expression Regulation, Neoplastic
- Humans
- Lung Diseases, Obstructive/complications
- Lung Diseases, Obstructive/drug therapy
- Lung Diseases, Obstructive/genetics
- Lung Diseases, Obstructive/metabolism
- Lung Neoplasms/complications
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Mesenchymal Stem Cells/pathology
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Pulmonary Emphysema/complications
- Pulmonary Emphysema/drug therapy
- Pulmonary Emphysema/genetics
- Pulmonary Emphysema/metabolism
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
Collapse
Affiliation(s)
- Vivek Mittal
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA.
- Department of Cardiothoracic Surgery, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA.
- Neuberger Berman Lung Cancer Research Center, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA.
| | - Tina El Rayes
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cardiothoracic Surgery, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA
- Neuberger Berman Lung Cancer Research Center, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA
- Weill Cornell Graduate School of Medical Sciences, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA
| | - Navneet Narula
- Department of Pathology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA
| | - Timothy E McGraw
- Department of Cardiothoracic Surgery, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA
- Neuberger Berman Lung Cancer Research Center, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA
- Department of Biochemistry, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA
| | - Nasser K Altorki
- Department of Cardiothoracic Surgery, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA
- Neuberger Berman Lung Cancer Research Center, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology, New York University School of Medicine, 566 First Avenue, New York, NY, 10016, USA.
| |
Collapse
|
38
|
Hypoxia inducible factor (HIF)-2α accelerates disease progression in mouse models of leukemia and lymphoma but is not a poor prognosis factor in human AML. Leukemia 2015; 29:2075-85. [DOI: 10.1038/leu.2015.102] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Revised: 03/12/2015] [Accepted: 03/30/2015] [Indexed: 12/15/2022]
|
39
|
Piao C, Cai L, Qiu S, Jia L, Song W, Du J. Complement 5a Enhances Hepatic Metastases of Colon Cancer via Monocyte Chemoattractant Protein-1-mediated Inflammatory Cell Infiltration. J Biol Chem 2015; 290:10667-76. [PMID: 25739439 DOI: 10.1074/jbc.m114.612622] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Indexed: 01/02/2023] Open
Abstract
Complement 5a (C5a), a potent immune mediator generated by complement activation, promotes tumor growth; however, its role in tumor metastasis remains unclear. We demonstrate that C5a contributes to tumor metastases by modulating tumor inflammation in hepatic metastases of colon cancer. Colon cancer cell lines generate C5a under serum-free conditions, and C5a levels increase over time in a murine syngeneic colon cancer hepatic metastasis model. Furthermore, in the absence of C5a receptor or upon pharmacological inhibition of C5a production with an anti-C5 monoclonal antibody, tumor metastasis is severely impaired. A lack of C5a receptor in colon cancer metastatic foci reduces the infiltration of macrophages, neutrophils, and dendritic cells, and the role for C5a receptor on these cells were further verified by bone marrow transplantation experiments. Moreover, C5a signaling increases the expression of the chemokine monocyte chemoattractant protein-1 and the anti-inflammatory molecules arginase-1, interleukin 10, and transforming growth factor β, but is inversely correlated with the expression of pro-inflammatory molecules, which suggests a mechanism for the role of C5a in the inflammatory microenvironment required for tumor metastasis. Our results indicate a new and potentially promising therapeutic application of complement C5a inhibitor for the treatment of malignant tumors.
Collapse
Affiliation(s)
- Chunmei Piao
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University, Beijing 100029 and The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Lun Cai
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University, Beijing 100029 and
| | - Shulan Qiu
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University, Beijing 100029 and
| | - Lixin Jia
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University, Beijing 100029 and
| | - Wenchao Song
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University, Beijing 100029 and
| | - Jie Du
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University, Beijing 100029 and The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China
| |
Collapse
|
40
|
Noh H, Hu J, Wang X, Xia X, Satelli A, Li S. Immune checkpoint regulator PD-L1 expression on tumor cells by contacting CD11b positive bone marrow derived stromal cells. Cell Commun Signal 2015; 13:14. [PMID: 25889536 PMCID: PMC4353689 DOI: 10.1186/s12964-015-0093-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/12/2015] [Indexed: 12/22/2022] Open
Abstract
Background Expression of programmed cell death ligand 1 (PD-L1) is an important process by which tumor cells suppress antitumor immunity in the tumor microenvironment. Bone marrow (BM)–derived immune cells are an important component of the tumor microenvironment. However, the link between PD-L1 induction on tumor cells and communication with BM cells is unknown. Results This study demonstrates that BM cells have a direct effect in inducing PD-L1 expression on tumor cells, which contributes to the tumor cells’ drug resistance. This novel discovery was revealed using a co-incubation system with BM cells and tumor cells. BM cells from wild-type C57BL6 mice and the immune-deficient mouse strains B-cell−/−, CD28−/−, perforin−/−, and Rag2−/− but not CD11b−/− dramatically increased the expression of tumor cell surface PD-L1. This PD-L1 induction was dependent on CD11b-positive BM cells through direct contact with tumor cells. Furthermore, p38 signaling was activated in tumor cells after co-incubation with BM cells, whereas the expression of PD-L1 was remarkably decreased after co-culture of cells treated with a p38 inhibitor. The increase in PD-L1 induced by BM cell co-culture protected tumor cells from drug-induced apoptosis. Conclusions PD-L1 expression is increased on tumor cells by direct contact with BM-derived CD11b-positive cells through the p38 signaling pathway. PD-L1 may play an important role in drug resistance, which often causes failure of the antitumor response. Electronic supplementary material The online version of this article (doi:10.1186/s12964-015-0093-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hyangsoon Noh
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Jiemiao Hu
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Xiaohong Wang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Xueqing Xia
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Arun Satelli
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Shulin Li
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
41
|
The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat Cell Biol 2014; 16:717-27. [PMID: 25082194 DOI: 10.1038/ncb3015] [Citation(s) in RCA: 715] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent pre-clinical and clinical research has provided evidence that cancer progression is driven not only by a tumour's underlying genetic alterations and paracrine interactions within the tumour microenvironment, but also by complex systemic processes. We review these emerging paradigms of cancer pathophysiology and discuss how a clearer understanding of systemic regulation of cancer progression could guide development of new therapeutic modalities and efforts to prevent disease relapse following initial diagnosis and treatment.
Collapse
|
42
|
Vail ME, Murone C, Tan A, Hii L, Abebe D, Janes PW, Lee FT, Baer M, Palath V, Bebbington C, Yarranton G, Llerena C, Garic S, Abramson D, Cartwright G, Scott AM, Lackmann M. Targeting EphA3 inhibits cancer growth by disrupting the tumor stromal microenvironment. Cancer Res 2014; 74:4470-81. [PMID: 25125683 DOI: 10.1158/0008-5472.can-14-0218] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Eph receptor tyrosine kinases are critical for cell-cell communication during normal and oncogenic tissue patterning and tumor growth. Somatic mutation profiles of several cancer genomes suggest EphA3 as a tumor suppressor, but its oncogenic expression pattern and role in tumorigenesis remain largely undefined. Here, we report unexpected EphA3 overexpression within the microenvironment of a range of human cancers and mouse tumor xenografts where its activation inhibits tumor growth. EphA3 is found on mouse bone marrow-derived cells with mesenchymal and myeloid phenotypes, and activation of EphA3(+)/CD90(+)/Sca1(+) mesenchymal/stromal cells with an EphA3 agonist leads to cell contraction, cell-cell segregation, and apoptosis. Treatment of mice with an agonistic α-EphA3 antibody inhibits tumor growth by severely disrupting the integrity and function of newly formed tumor stroma and microvasculature. Our data define EphA3 as a novel target for selective ablation of the tumor microenvironment and demonstrate the potential of EphA3 agonists for anticancer therapy.
Collapse
Affiliation(s)
- Mary E Vail
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia.
| | - Carmel Murone
- Ludwig Institute for Cancer Research, Melbourne, Victoria, Australia
| | - April Tan
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Linda Hii
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Degu Abebe
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Peter W Janes
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Fook-Thean Lee
- Ludwig Institute for Cancer Research, Melbourne, Victoria, Australia
| | - Mark Baer
- KaloBios Pharmaceuticals, Inc., South San Francisco, California
| | - Varghese Palath
- KaloBios Pharmaceuticals, Inc., South San Francisco, California
| | | | | | - Carmen Llerena
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Slavisa Garic
- Faculty of Information Technology, Monash University, Clayton, Victoria, Australia
| | - David Abramson
- Faculty of Information Technology, Monash University, Clayton, Victoria, Australia
| | - Glenn Cartwright
- Ludwig Institute for Cancer Research, Melbourne, Victoria, Australia
| | - Andrew M Scott
- Ludwig Institute for Cancer Research, Melbourne, Victoria, Australia. Faculty of Medicine, University of Melbourne, Victoria, Australia.
| | - Martin Lackmann
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia.
| |
Collapse
|
43
|
CSF1-ETS2-induced microRNA in myeloid cells promote metastatic tumor growth. Oncogene 2014; 34:3651-61. [PMID: 25241894 PMCID: PMC4369473 DOI: 10.1038/onc.2014.294] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/15/2014] [Accepted: 08/04/2014] [Indexed: 01/02/2023]
Abstract
Metastasis of solid tumors is associated with poor prognosis and bleak survival rates. Tumor infiltrating myeloid cells (TIMs) are known to promote metastasis but the mechanisms underlying their collaboration with tumor cells remain unknown. Here we report an oncogenic role for microRNA in driving M2 reprogramming in TIMs, characterized by the acquisition of pro-tumor and pro-angiogenic properties. The expression of miR-21, miR-29a, miR-142-3p and miR-223 increased in myeloid cells during tumor progression in mouse models of breast cancer and melanoma metastasis. Further, we show that these miRs are regulated by the CSF1-ETS2 pathway in macrophages. A loss of function approach utilizing selective depletion of the microRNA processing enzyme Dicer in mature myeloid cells blocks angiogenesis and metastatic tumor growth. Ectopic expression of miR-21 and miR-29a promotes angiogenesis and tumor cell proliferation through the down-regulation of anti-angiogenic genes such as Col4a2, Spry1 and Timp3 whereas knockdown of the miRs impedes these processes. miR-21 and miR-29a are expressed in Csf1r+ myeloid cells associated with human metastatic breast cancer and levels of these miRs in CD115+ non-classical monocytes correlates with metastatic tumor burden in patients. Taken together, our results suggest that miR-21 and miR-29a are essential for the pro-tumor functions of myeloid cells and the CSF1-ETS2 pathway upstream of the miRs serves as an attractive therapeutic target for the inhibition of M2 remodeling of macrophages during malignancy. In addition, miR-21 and miR-29a in circulating myeloid cells may potentially serve as biomarkers to measure therapeutic efficacy of targeted therapies for CSF1 signaling.
Collapse
|
44
|
Bruun J, Kolberg M, Nesland JM, Svindland A, Nesbakken A, Lothe RA. Prognostic Significance of β-Catenin, E-Cadherin, and SOX9 in Colorectal Cancer: Results from a Large Population-Representative Series. Front Oncol 2014; 222:1-15. [PMID: 24904831 DOI: 10.1002/path.2727] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Robust biomarkers that can precisely stratify patients according to treatment needs are in great demand. The literature is inconclusive for most reported prognostic markers for colorectal cancer (CRC). Hence, adequately reported studies in large representative series are necessary to determine their clinical potential. We investigated the prognostic value of three Wnt signaling-associated proteins, β-catenin, E-cadherin, and SOX9, in a population-representative single-hospital series of 1290 Norwegian CRC patients by performing immunohistochemical analyses of each marker using the tissue microarray technology. Loss of membranous or cytosolic β-catenin and loss of cytosolic E-cadherin protein expression were significantly associated with reduced 5-year survival in 903 patients who underwent major resection (722 evaluable tissue cores) independently of standard clinicopathological high-risk parameters. Pre-specified subgroup analyses demonstrated particular effect for stage IV patients for β-catenin membrane staining (P = 0.018; formal interaction test P = 0.025). Among those who underwent complete resection (714 patients, 568 evaluable), 5-year time-to-recurrence analyses were performed, and stage II patients with loss of cytosolic E-cadherin were identified as an independent high-risk subgroup (P = 0.020, formal interaction test was not significant). Nuclear β-catenin and SOX9 protein, regardless of intracellular location, were not associated with prognosis. In conclusion, the protein expression level of membranous or cytosolic β-catenin and E-cadherin predicts CRC patient subgroups with inferior prognosis.
Collapse
Affiliation(s)
- Jarle Bruun
- Department for Cancer Prevention, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital , Oslo , Norway ; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo , Oslo , Norway
| | - Matthias Kolberg
- Department for Cancer Prevention, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital , Oslo , Norway ; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo , Oslo , Norway
| | - Jahn M Nesland
- Department of Pathology, Oslo University Hospital , Oslo , Norway
| | - Aud Svindland
- Department of Pathology, Oslo University Hospital , Oslo , Norway ; Faculty of Medicine, University of Oslo , Oslo , Norway
| | - Arild Nesbakken
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo , Oslo , Norway ; Faculty of Medicine, University of Oslo , Oslo , Norway ; Department of Gastrointestinal Surgery, Aker Hospital, Oslo University Hospital , Oslo , Norway
| | - Ragnhild A Lothe
- Department for Cancer Prevention, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital , Oslo , Norway ; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo , Oslo , Norway ; Department of Molecular Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo , Oslo , Norway
| |
Collapse
|
45
|
Inhibitor of DNA binding 1 as a secreted angiogenic transcription factor in rheumatoid arthritis. Arthritis Res Ther 2014; 16:R68. [PMID: 24620998 PMCID: PMC4060463 DOI: 10.1186/ar4507] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 03/04/2014] [Indexed: 12/17/2022] Open
Abstract
Introduction Rheumatoid arthritis (RA) is characterized by enhanced blood vessel development in joint synovium. This involves the recruitment of endothelial progenitor cells (EPCs), allowing for de novo vessel formation and pro-inflammatory cell infiltration. Inhibitor of DNA Binding 1 (Id1) is a transcription factor characteristic of EPCs that influences cell maturation. Method Enzyme-linked immunosorbant assay (ELISA) and polymerase chain reaction (PCR) were used to examine Id1 levels in synovial fluid (SF) and endothelial cells (ECs), respectively. Immunohistology was used to determine the expression of Id1 in synovial tissue (ST). Human dermal microvascular EC (HMVEC) migration and tube forming assays were used to determine if recombinant human Id1 (rhuId1) and/or RA SF immunodepleted Id1 showed angiogenic activity. We also utilized the RA ST severe combined immunodeficient (SCID) mouse chimera to examine if Id1 recruits EPCs to RA synovium. Results ST samples immunostained for Id1 showed heightened expression in RA compared to osteoarthritis (OA) and normal (NL) ST. By immunofluorescence staining, we found significantly more Id1 in RA compared to OA and NL vasculature, showing that Id1 expressing cells, and therefore EPCs, are most active in vascular remodeling in the RA synovium. We also detected significantly more Id1 in RA compared to OA and other arthritis SFs by ELISA, which correlates highly with Chemokine (C-X-C motif) ligand 16 (CXCL16) levels. In vitro chemotaxis assays showed that Id1 is highly chemotactic for HMVECs and can be attenuated by inhibition of Nuclear Factor κB and phosphoinositide 3-kinase. Using in vitro Matrigel assays, we found that HMVECs form tubes in response to rhuId1 and that Id1 immunodepleted from RA SF profoundly decreases tube formation in Matrigel in vitro. PCR showed that Id1 mRNA could be up-regulated in EPCs compared to HMVECs in response to CXCL16. Finally, using the K/BxN serum induced arthritis model, we found that EC CXCR6 correlated with Id1 expression by immunohistochemistry. Conclusions We conclude that Id1 correlates highly with CXCL16 expression, EPC recruitment, and blood vessel formation in the RA joint, and that Id1 is potently angiogenic and can be up-regulated in EPCs by CXCL16.
Collapse
|
46
|
Signal transduction in tumor angiogenesis. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
47
|
Is carcinoma a mesenchymal disease? The role of the stromal microenvironment in carcinogenesis. Pathology 2013; 45:371-81. [PMID: 23594691 DOI: 10.1097/pat.0b013e328360b600] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Most research into the biology of carcinoma has focused on the epithelial cells therein; the inherent assumption has been that the tumour arises from epithelial cells 'gone bad', and that the surrounding stroma is simply an 'innocent bystander'. However, there is increasing evidence that there is a complex interplay between tumour cells and their surrounding microenvironment, and that the latter may be just as important in determining the development and clinical behaviour of a given tumour. Similarly, traditional oncological practice has been predominantly aimed at a perceived ideal goal of killing all the tumour epithelial cells, with only a few recently developed therapies seeking to affect other components (such as tumour vasculature); but identifying stromal factors involved in tumour growth and survival may well lead to the development of novel therapies. This review examines current understanding of the interplay between tumour epithelial cells and their microenvironment, and enumerates various stromal factors which appear to play a role in tumour progression and/or metastasis.
Collapse
|
48
|
Feng G, Jiang F, Pan C, Pu C, Huang H, Li G. Quantification of peripheral blood CD133 mRNA in identifying metastasis and in predicting recurrence of patients with clear cell renal cell carcinoma. Urol Oncol 2013; 32:44.e9-14. [PMID: 24054866 DOI: 10.1016/j.urolonc.2013.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 06/06/2013] [Accepted: 06/11/2013] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To investigate whether CD133 messenger ribonucleic acid (mRNA) could provide useful information to identify metastasis or predict recurrence in patients with clear cell renal cell carcinoma (cRCC). METHODS AND MATERIALS This study included 86 patients with cRCC and 30 healthy controls. Real-time reverse transcriptase-polymerase chain reaction was used to quantify CD133 mRNA in peripheral blood mononuclear cells before nephrectomy. RESULTS The average CD133 mRNA in patients with metastatic cRCC (1.546 ± 0.291) was significantly higher than that in those with localized cRCC (1.034 ± 0.316, P = 0.022) or in controls (0.042 ± 0.028, P = 0.001). Metastasis could be identified with a sensitivity of 82.6% at specificity of 69.8% by CD133 mRNA. Among patients with localized cRCC, there was a significant difference in CD133 mRNA between the patients with recurrence (1.136 ± 0.127) and without recurrence (1.010 ± 0.091, P = 0.047). Recurrence could be identified with a sensitivity of 75.0% at specificity of 61.8%. Patients with a higher CD133 mRNA had a significantly higher recurrence rate than those with a low CD133 mRNA (P = 0.019). CONCLUSIONS CD133 mRNA can be useful for identifying metastasis, predicting recurrence, and stratifying the patients into different risk groups for possible adjuvant treatment.
Collapse
Affiliation(s)
- Gang Feng
- Clinical Genetics Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Feng Jiang
- Department of Ultrasound, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Caiming Pan
- Department of Urology, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Chun Pu
- Clinical Genetics Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Houbao Huang
- Department of Urology, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China.
| | - Guorong Li
- Department of Urology, North Hospital, CHU of Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
49
|
Abstract
Human endothelial progenitor cells (EPCs) have been generally defined as circulating cells that express a variety of cell surface markers similar to those expressed by vascular endothelial cells, adhere to endothelium at sites of hypoxia/ischemia, and participate in new vessel formation. Although no specific marker for an EPC has been identified, a panel of markers has been consistently used as a surrogate marker for cells displaying the vascular regenerative properties of the putative EPC. However, it is now clear that a host of hematopoietic and vascular endothelial subsets display the same panel of antigens and can only be discriminated by an extensive gene expression analysis or use of a variety of functional assays that are not often applied. This article reviews our current understanding of the many cell subsets that constitute the term EPC and provides a concluding perspective as to the various roles played by these circulating or resident cells in vessel repair and regeneration in human subjects.
Collapse
Affiliation(s)
- Mervin C Yoder
- Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
50
|
Caiado F, Carvalho T, Rosa I, Remédio L, Costa A, Matos J, Heissig B, Yagita H, Hattori K, da Silva JP, Fidalgo P, Pereira AD, Dias S. Bone marrow-derived CD11b+Jagged2+ cells promote epithelial-to-mesenchymal transition and metastasization in colorectal cancer. Cancer Res 2013; 73:4233-46. [PMID: 23722542 DOI: 10.1158/0008-5472.can-13-0085] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Timely detection of colorectal cancer metastases may permit improvements in their clinical management. Here, we investigated a putative role for bone marrow-derived cells in the induction of epithelial-to-mesenchymal transition (EMT) as a marker for onset of metastasis. In ectopic and orthotopic mouse models of colorectal cancer, bone marrow-derived CD11b(Itgam)(+)Jagged2 (Jag2)(+) cells infiltrated primary tumors and surrounded tumor cells that exhibited diminished expression of E-cadherin and increased expression of vimentin, 2 hallmarks of EMT. In vitro coculture experiments showed that the bone marrow-derived CD11b(+)Jag2(+) cells induced EMT through a Notch-dependent pathway. Using neutralizing antibodies, we imposed a blockade on CD11b(+) cells' recruitment to tumors, which decreased the tumor-infiltrating CD11b(+)Jag2(+) cell population of interest, decreasing tumor growth, restoring E-cadherin expression, and delaying EMT. In support of these results, we found that peripheral blood levels of CD11b(+)Jag2(+) cells in mouse models of colorectal cancer and in a cohort of untreated patients with colorectal cancer were indicative of metastatic disease. In patients with colorectal cancer, the presence of circulating CD11b(+)Jag2(+) cells was accompanied by loss of E-cadherin in the corresponding patient tumors. Taken together, our results show that bone marrow-derived CD11b(+)Jag2(+) cells, which infiltrate primary colorectal tumors, are sufficient to induce EMT in tumor cells, thereby triggering onset of metastasis. Furthermore, they argue that quantifying circulating CD11b(+)Jag2(+) cells in patients may offer an indicator of colorectal cancer progression to metastatic levels of the disease.
Collapse
Affiliation(s)
- Francisco Caiado
- Angiogenesis Lab, CIPM, Department of Gastroenterology, and Histopathology Unit, Portuguese Institute of Oncology, IPOLFG, EPE, Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Lisbon, and Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|