1
|
Arebro J, Lee CM, Bennewith KL, Garnis C. Cancer-Associated Fibroblast Heterogeneity in Malignancy with Focus on Oral Squamous Cell Carcinoma. Int J Mol Sci 2024; 25:1300. [PMID: 38279300 PMCID: PMC10816981 DOI: 10.3390/ijms25021300] [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: 12/01/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/28/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) remains an understudied and significant global cancer killer and dismal survival rates have not changed in decades. A better understanding of the molecular basis of OSCC progression and metastasis is needed to develop new approaches for treating this disease. The supportive network surrounding cancer tumor cells known as the tumor microenvironment (TME) has gained increasing interest lately since it performs essential protumorigenic functions. Cancer-associated fibroblasts (CAFs) are one of the main cell types in the TME and are known to play a key role in influencing the biological behavior of tumors. CAFs present a heterogeneity both in phenotype as well as functions, leading to the suggestion of different CAF subtypes in several cancer forms. The task to subtype CAFs in OSCC has, however, just begun, and there is today no united way of subtyping CAFs in this disease. This review aims to define the features of CAFs and to summarize CAF subtype research in malignancy with focus on OSCC including aspects as disease prognosis and therapeutic opportunities.
Collapse
Affiliation(s)
- Julia Arebro
- Department of Interdisciplinary Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (C.-M.L.); (K.L.B.); (C.G.)
- Division of ENT Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14186 Stockholm, Sweden
- Department of ENT Diseases, Karolinska University Hospital, 14186 Stockholm, Sweden
| | - Che-Min Lee
- Department of Interdisciplinary Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (C.-M.L.); (K.L.B.); (C.G.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Kevin L. Bennewith
- Department of Interdisciplinary Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (C.-M.L.); (K.L.B.); (C.G.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5Z 1L3, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | - Cathie Garnis
- Department of Interdisciplinary Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (C.-M.L.); (K.L.B.); (C.G.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V5Z 1L3, Canada
- Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
2
|
Qiu H, Zhang X, Qi J, Zhang J, Tong Y, Li L, Fu L, Qin YR, Guan X, Zhang L. Identification and characterization of FGFR2+ hematopoietic stem cell-derived fibrocytes as precursors of cancer-associated fibroblasts induced by esophageal squamous cell carcinoma. J Exp Clin Cancer Res 2022; 41:240. [PMID: 35941662 PMCID: PMC9358838 DOI: 10.1186/s13046-022-02435-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/06/2022] [Indexed: 11/28/2022] Open
Abstract
Background Cancer-associated fibroblast (CAF) is an ideal target for cancer treatment. Recent studies have focused on eliminating CAFs and their effects by targeting their markers or blocking individual CAF-secreted factors. However, these strategies have been limited by their specificity for targeting CAFs and effectiveness in blocking widespread influence of CAFs. To optimize CAF-targeted therapeutic strategies, we tried to explore the molecular mechanisms of CAF generation in this study. Methods Using FGFR2 as a tracing marker, we identified a novel origin of CAFs in esophageal squamous cell carcinoma (ESCC). Furthermore, we successfully isolated CAF precursors from peripheral blood of ESCC patients and explored the mechanisms underlying their expansion, recruitment, and differentiation via RNA-sequencing and bioinformatics analysis. The mechanisms were further verified by using different models both in vitro and in vivo. Results We found that FGFR2+ hematopoietic stem cell (HSC)-derived fibrocytes could be induced by ESCC cells, recruited into tumor xenografts, and differentiated into functional CAFs. They were mobilized by cancer-secreted FGF2 and recruited into tumor sites via the CXCL12/CXCR4 axis. Moreover, they differentiated into CAFs through the activation of YAP-TEAD complex, which is triggered by directly contracting with tumor cells. FGF2 and CXCR4 neutralizing antibodies could effectively block the mobilization and recruitment process of FGFR2+ CAFs. The YAP-TEAD complex-based mechanism hold promise for locally activation of genetically encoded therapeutic payloads at tumor sites. Conclusions We identified a novel CAF origin and systematically studied the process of mobilization, recruitment, and maturation of CAFs in ESCC under the guidance of tumor cells. These findings give rise to new approaches that target CAFs before their incorporation into tumor stroma and use CAF-precursors as cellular vehicles to target tumor cells. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02435-w.
Collapse
|
3
|
Papait A, Romoli J, Stefani FR, Chiodelli P, Montresor MC, Agoni L, Silini AR, Parolini O. Fight the Cancer, Hit the CAF! Cancers (Basel) 2022; 14:cancers14153570. [PMID: 35892828 PMCID: PMC9330284 DOI: 10.3390/cancers14153570] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary In the last 20 years, the tumor microenvironment (TME) has raised an increasing interest from the therapeutic point of view. Indeed, different strategies targeting either the endothelial or the immune component have been implemented. Furthermore, cancer-associated fibroblasts (CAF) have attracted even more interest due to their ability to prime the TME in order to favor tumor progression and metastasis. This current review provides a comprehensive overview on the latest discoveries regarding CAF, more specifically on their complex characterization and on preclinical studies and clinical trials that target CAF within the TME. Abstract The tumor microenvironment (TME) is comprised of different cellular components, such as immune and stromal cells, which co-operate in unison to promote tumor progression and metastasis. In the last decade, there has been an increasing focus on one specific component of the TME, the stromal component, often referred to as Cancer-Associated Fibroblasts (CAF). CAF modulate the immune response and alter the composition of the extracellular matrix with a decisive impact on the response to immunotherapies and conventional chemotherapy. The most recent publications based on single-cell analysis have underlined CAF heterogeneity and the unique plasticity that strongly impact the TME. In this review, we focus not only on the characterization of CAF based on the most recent findings, but also on their impact on the immune system. We also discuss clinical trials and preclinical studies where targeting CAF revealed controversial results. Therefore, future efforts should focus on understanding the functional properties of individual subtypes of CAF, taking into consideration the peculiarities of each pathological context.
Collapse
Affiliation(s)
- Andrea Papait
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (J.R.)
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy
| | - Jacopo Romoli
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (J.R.)
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (F.R.S.); (P.C.); (A.R.S.)
| | - Francesca Romana Stefani
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (F.R.S.); (P.C.); (A.R.S.)
| | - Paola Chiodelli
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (F.R.S.); (P.C.); (A.R.S.)
| | | | - Lorenzo Agoni
- Obstetrics and Gynecology Unit, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy;
| | - Antonietta Rosa Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (F.R.S.); (P.C.); (A.R.S.)
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (J.R.)
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy
- Correspondence: ; Tel.: +39-0630154464
| |
Collapse
|
4
|
Hachiya K, Masuya M, Kuroda N, Yoneda M, Tsuboi J, Nagaharu K, Nishimura K, Shiotani T, Ohishi K, Tawara I, Katayama N. Irbesartan, an angiotensin II type 1 receptor blocker, inhibits colitis-associated tumourigenesis by blocking the MCP-1/CCR2 pathway. Sci Rep 2021; 11:19943. [PMID: 34620946 PMCID: PMC8497524 DOI: 10.1038/s41598-021-99412-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/20/2021] [Indexed: 02/07/2023] Open
Abstract
The introduction of anti-inflammatory therapies has enabled substantial improvement of disease activity in patients with inflammatory bowel diseases (IBD). However, IBD can lead to serious complications such as intestinal fibrosis and colorectal cancer. Therefore, novel therapies reducing the development of these complications are needed. Angiotensin II (Ang II) promotes tissue inflammation by stimulating the production of monocyte chemoattractant protein-1 (MCP-1) or proinflammatory cytokines. It plays a pivotal role in IBD progression. Although blockade of Ang II has been reported to ameliorate experimental colitis and reduce colorectal cancer risk, the cellular and molecular mechanisms remain poorly understood. Our previous work showed that irbesartan, an Ang II type 1 receptor blocker, reduced the number of C-C chemokine receptor 2-positive (CCR2+) monocytic cells in the inflamed pancreas. This study aimed to investigate the possible antifibrotic and antitumour effects of irbesartan using the azoxymethane/dextran sodium sulphate mouse model. Irbesartan suppressed MCP-1 production and the accumulation of Ly6C+CCR2+ monocytes and fibrocytes in the inflamed colon, downregulated the expression of type 1 collagen and matrix metalloproteinase 9 and inhibited the development of intestinal fibrosis and tumours. Our observations suggest that blocking the MCP-1/CCR2 pathway using irbesartan might be beneficial in preventing colitis-associated colon tumours.
Collapse
Affiliation(s)
- Kensuke Hachiya
- Department of Haematology and Oncology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Masahiro Masuya
- Department of Haematology and Oncology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan.
- Course of Nursing Science, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
| | - Naoki Kuroda
- Department of Gastroenterology, Saiseikai Matsusaka General Hospital, Matsusaka, Mie, 515-8557, Japan
| | - Misao Yoneda
- Department of Clinical Nutrition Medical Technology Course, Suzuka University of Medical Science, Suzuka, Mie, 510-0293, Japan
| | - Junya Tsuboi
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Keiki Nagaharu
- Department of Haematology and Oncology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Komei Nishimura
- Department of Haematology and Oncology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Takuya Shiotani
- Department of Haematology and Oncology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Kohshi Ohishi
- Department of Transfusion Medicine and Cell Therapy, Mie University Hospital, Tsu, Mie, 514-8507, Japan
| | - Isao Tawara
- Department of Haematology and Oncology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Naoyuki Katayama
- Department of Haematology and Oncology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
- Faculty of Nursing, Suzuka University of Medical Science, Suzuka, Mie, 513-8670, Japan
| |
Collapse
|
5
|
Cancer-associated fibroblasts: overview, progress, challenges, and directions. Cancer Gene Ther 2021; 28:984-999. [PMID: 33712707 DOI: 10.1038/s41417-021-00318-4] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/12/2021] [Accepted: 02/25/2021] [Indexed: 01/30/2023]
Abstract
Tumors are one of the main causes of death in humans. The development of safe and effective methods for early diagnosis and treatment of tumors is a difficult problem that needs to be solved urgently. It is well established that the occurrence of tumors involves complex biological mechanisms, and the tumor microenvironment (TME) plays an important role in regulating the biological behavior of tumors. Cancer-associated fibroblasts (CAFs) are a group of activated fibroblasts with significant heterogeneity and plasticity in the tumor microenvironment. They secrete a variety of active factors to regulate tumor occurrence, development, metastasis, and therapeutic resistance. Although most studies suggest that CAFs have significant tumor-promoting functions, some evidence indicates that they may have certain tumor-suppressive functions in the early stage of tumors. Current research on CAFs continues to face many challenges, and the heterogeneity of their origin, phenotype, and function is a major difficulty and hot spot. To provide new perspectives for the research on CAFs and tumor diagnosis and treatment, this review summarizes the definition, origin, biomarkers, generation mechanism, functions, heterogeneity, plasticity, subpopulations, pre-metastasis niches (PMN), immune microenvironment, and targeted therapy of CAFs, describes the research progress and challenges, and proposes possible future research directions based on existing reports.
Collapse
|
6
|
Riley HJ, Kelly RR, Van Laer AO, Neff LS, Dasgupta S, Baicu CF, McDonald LT, LaRue AC, Zile MR, Bradshaw AD. SPARC production by bone marrow-derived cells contributes to myocardial fibrosis in pressure overload. Am J Physiol Heart Circ Physiol 2021; 320:H604-H612. [PMID: 33306449 PMCID: PMC8082795 DOI: 10.1152/ajpheart.00552.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022]
Abstract
In human heart failure and in murine hearts with left-ventricular pressure overload (LVPO), increases in fibrosis are associated with increases in myocardial stiffness. Secreted protein acidic and rich in cysteine (SPARC) is shown to be necessary for both cardiac fibrosis and increases in myocardial stiffness in response to LVPO; however, cellular sources of cardiac SPARC are incompletely defined. Irradiation and bone marrow transfer were undertaken to test the hypothesis that SPARC expression by bone marrow-derived cells is an important mediator of fibrosis in LVPO. In recipient SPARC-null mice transplanted with donor wild-type (WT) bone marrow and subjected to LVPO, levels of fibrosis similar to that of WT mice were found despite the lack of SPARC expression by resident cells. In recipient WT mice with donor SPARC-null bone marrow, significantly less fibrosis versus that of WT mice was found despite the expression of SPARC by resident cells. Increases in myocardial stiffness followed a similar pattern to that of collagen deposition. Myocardial macrophages were significantly reduced in SPARC-null mice with LVPO versus that of WT mice. Recipient SPARC-null mice transplanted with donor WT bone marrow exhibited an increase in cardiac macrophages versus that of SPARC-null LVPO and donor WT mice with recipient SPARC-null bone marrow. Expression of vascular cellular adhesion molecule (VCAM), a previously identified binding partner of SPARC, was assessed in all groups and with the exception of WT mice, increases in VCAM immunoreactivity with LVPO were observed. However, no differences in VCAM expression between bone marrow transplant groups were noted. In conclusion, SPARC expression by bone marrow-derived cells was critical for fibrotic deposition of collagen and influenced the expansion of myocardial macrophages in response to LVPO.NEW & NOTEWORTHY Myocardial fibrosis and the resultant increases in LV and myocardial stiffness represent pivotal consequences of chronic pressure overload (PO). In this study, a murine model of cardiac fibrosis induced by PO was used to demonstrate a critical function of SPARC in bone marrow-derived cells that drives cardiac fibrosis and increases in cardiac macrophages.
Collapse
Affiliation(s)
- Hannah J Riley
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Ryan R Kelly
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - An O Van Laer
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Lily S Neff
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Shaoni Dasgupta
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Catalin F Baicu
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Lindsay T McDonald
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Amanda C LaRue
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Michael R Zile
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Amy D Bradshaw
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
7
|
Xu Z, Sun Y, Wei Z, Jiang J, Xu J, Liu P. Suppression of CXCL-1 Could Restore Necroptotic Pathway in Chronic Lymphocytic Leukemia. Onco Targets Ther 2020; 13:6917-6925. [PMID: 32764983 PMCID: PMC7371606 DOI: 10.2147/ott.s256993] [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: 04/18/2020] [Accepted: 06/17/2020] [Indexed: 01/18/2023] Open
Abstract
Purpose To clarify the role of different cytokines and selenite in the defective necroptotic pathway of chronic lymphocytic leukemia (CLL). Patients and Methods We randomly collected the peripheral blood samples of 11 untreated CLL patients and 10 healthy volunteers, and then separated B lymphocytes from peripheral blood. Then, real-time polymerase chain reaction (PCR), enzyme-linked immunosorbent assay (ELISA) and Western Blot were performed to detect the expression of different cytokines, including CXC-motif chemokine ligand 1 (CXCL-1). Finally, we used flow cytometry to analyze the percentage of surviving cells to figure out whether CLL cells or normal B lymphocytes underwent necroptosis. Results 1) The high expression of CXCL-1 was seen in CLL cells compared with normal B lymphocytes (p = 0.0001, adjusted p =0.0012); 2) The downregulation of CXCL-1 was shown in normal B lymphocytes after induction by TNF-α and z-VAD; 3) CLL cells could restore necroptosis induced by TNF-α and z-VAD after knockdown of CXCL-1; 4) The transcriptional and translational expression of LEF-1 were downregulated after the knockdown of CXCL-1 in CLL cells; 5. 3.2μM selenite could help CLL cells restore necroptosis (p = 0.0102) and inhibit the transcriptional and translational expression of CXCL-1. Conclusion CXCL-1 played an important role in the defective necroptosis of CLL cells and regulated the expression of LEF-1. Selenite could inhibit the expression of CXCL-1 and help CLL cells restore necroptosis together with TNF-α and z-VAD. Selenite might be the potential medication of CLL in the future.
Collapse
Affiliation(s)
- Zhao Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yifeng Sun
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Zheng Wei
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jifeng Jiang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jiadai Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
8
|
Rossi E, Zamarchi R. Single-Cell Analysis of Circulating Tumor Cells: How Far Have We Come in the -Omics Era? Front Genet 2019; 10:958. [PMID: 31681412 PMCID: PMC6811661 DOI: 10.3389/fgene.2019.00958] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022] Open
Abstract
Tumor cells detach from the primary tumor or metastatic sites and enter the peripheral blood, often causing metastasis. These cells, named Circulating Tumor Cells (CTCs), display the same spatial and temporal heterogeneity as the primary tumor. Since CTCs are involved in tumor progression, they represent a privileged window to disclose mechanisms of metastases, while -omic analyses at the single-cell level allow dissection of the complex relationships between the tumor subpopulations and the surrounding normal tissue. However, in addition to reporting the proof of concept that we can query CTCs to reveal tumor evolution throughout the continuum of treatment for early detection of resistance to therapy, the scientific literature has also been highlighting the disadvantages of CTCs, which hampers a routine use of this approach in clinical practice. To date, an increasing number of CTC technologies, as well as -omics methods, have been employed, mostly lacking strong comparative analyses. The rarity of CTCs also represents a major challenge, because there is no consensus regarding the minimal criteria necessary and sufficient to define an event as CTC; moreover, we cannot often compare data from of one study with that of another. Finally, the availability of an individual tumor profile undermines the traditional histology-based treatment. Applying molecular data for patient benefit implies a collective effort by biologists, bioengineers, and clinicians, to create tools to interpret molecular data and manage precision medicine in every single patient. Herein, we focus on the most recent findings in CTC −omics to learn how far we have come.
Collapse
Affiliation(s)
- Elisabetta Rossi
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy.,Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Rita Zamarchi
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| |
Collapse
|
9
|
High-Fat Diet Alters Immunogenic Properties of Circulating and Adipose Tissue-Associated Myeloid-Derived CD45 +DDR2 + Cells. Mediators Inflamm 2019; 2019:1648614. [PMID: 31015794 PMCID: PMC6421777 DOI: 10.1155/2019/1648614] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/04/2019] [Accepted: 01/15/2019] [Indexed: 12/26/2022] Open
Abstract
Chronic inflammation is evident in the adipose tissue and periphery of patients with obesity, as well as mouse models of obesity. T cell subsets in obese adipose tissue are skewed towards Th1- and Th17-associated phenotypes and their secreted cytokines contribute to obesity-associated inflammation. Our lab recently identified a novel, myeloid-derived CD45+DDR2+ cell subset that modulates T cell activity. The current study sought to determine how these myeloid-derived CD45+DDR2+ cells are altered in the adipose tissue and peripheral blood of preobese mice and how this population modulates T cell activity. C57BL/6 mice were fed with a diet high in milkfat (60%·kcal, HFD) ad libitum until a 20% increase in total body weight was reached, and myeloid-derived CD45+DDR2+ cells and CD4+ T cells in visceral adipose tissue (VAT), mammary gland-associated adipose tissue (MGAT), and peripheral blood (PB) were phenotypically analyzed. Also analyzed was whether mediators from MGAT-primed myeloid-derived CD45+DDR2+ cells stimulate normal CD4+ T cell cytokine production. A higher percentage of myeloid-derived CD45+DDR2+ cells expressed the activation markers MHC II and CD80 in both VAT and MGAT of preobese mice. CD4+ T cells were preferentially skewed towards Th1- and Th17-associated phenotypes in the adipose tissue and periphery of preobese mice. In vitro, MGAT from HFD-fed mice triggered myeloid-derived CD45+DDR2+ cells to induce CD4+ T cell IFN-γ and TNF-α production. Taken together, this study shows that myeloid-derived CD45+DDR2+ cells express markers of immune activation and suggests that they play an immune modulatory role in the adipose tissue of preobese mice.
Collapse
|
10
|
Liu FD, Tam K, Pishesha N, Poon Z, Van Vliet KJ. Improving hematopoietic recovery through modeling and modulation of the mesenchymal stromal cell secretome. Stem Cell Res Ther 2018; 9:268. [PMID: 30352620 PMCID: PMC6199758 DOI: 10.1186/s13287-018-0982-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 08/13/2018] [Accepted: 08/15/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Efficient and sustained hematopoietic recovery after hematopoietic stem cell or bone marrow transplantation is supported by paracrine signaling from specific subpopulations of mesenchymal stromal cells (MSCs). Here, we considered whether in vitro mechanopriming of human MSCs could be administered to predictively and significantly improve in vivo hematopoietic recovery after irradiation injury. METHODS First, we implemented regression modeling to identify eight MSC-secreted proteins that correlated strongly with improved rescue from radiation damage, including hematopoietic recovery, in a murine model of hematopoietic failure. Using these partial least squares regression (PLSR) model parameters, we then predicted recovery potential of MSC populations that were culture expanded on substrata of varying mechanical stiffness. Lastly, we experimentally validated these predictions using an in vitro co-culture model of hematopoiesis and using new in vivo experiments for the same irradiation injury model used to generate survival predictions. RESULTS MSCs grown on the least stiff (elastic moduli ~ 1 kPa) of these polydimethylsiloxane (PDMS) substrata secreted high concentrations of key proteins identified in regression modeling, at concentrations comparable to those secreted by minor subpopulations of MSCs shown previously to be effective in supporting such radiation rescue. We confirmed that these MSCs expanded on PDMS could promote hematopoiesis in an in vitro co-culture model with hematopoietic stem and progenitor cells (HSPCs). Further, MSCs cultured on PDMS of highest stiffness (elastic moduli ~ 100 kPa) promoted expression of CD123+ HSPCs, indicative of myeloid differentiation. Systemic administration of mechanoprimed MSCs resulted in improved mouse survival and weight recovery after bone marrow ablation, as compared with both standard MSC expansion on stiffer materials and with biophysically sorted MSC subpopulations. Additionally, we observed recovery of white blood cells, platelets, and red blood cells, indicative of complete recovery of all hematopoietic lineages. CONCLUSIONS These results demonstrate that computational techniques to identify MSC biomarkers can be leveraged to predict and engineer therapeutically effective MSC phenotypes defined by mechanoprimed secreted factors, for translational applications including hematopoietic recovery.
Collapse
Affiliation(s)
- Frances D. Liu
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 USA
- Biosystems and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore, 138602 Singapore
| | - Kimberley Tam
- Biosystems and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore, 138602 Singapore
| | - Novalia Pishesha
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 USA
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02139 USA
| | - Zhiyong Poon
- Biosystems and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore, 138602 Singapore
| | - Krystyn J. Van Vliet
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 USA
- Biosystems and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore, 138602 Singapore
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 USA
| |
Collapse
|
11
|
Wang T, Zhan Q, Peng X, Qiu Z, Zhao T. CCL2 influences the sensitivity of lung cancer A549 cells to docetaxel. Oncol Lett 2018; 16:1267-1274. [PMID: 30061946 PMCID: PMC6063033 DOI: 10.3892/ol.2018.8769] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 03/09/2018] [Indexed: 01/09/2023] Open
Abstract
Lung cancer is one of the most common malignant tumor types globally. Acquisition of chemoresistance in lung cancer cells is the primary cause of chemotherapy failure. Inflammatory chemokine C-C motif chemokine ligand 2 (CCL2) has been reported to be involved in the progression of cancer and drug resistance. However, its function in docetaxel (DTX) resistance of lung cancer remains unclear. In the present study, the mechanism underlying DTX-induced drug resistance was investigated. Reverse transcription-quantitative polymerase chain reaction and western blot analysis revealed that DTX treatment increased the mRNA and protein expression of CCL2 in lung cancer A549 cells. CCL2 was knocked down by small interfering RNA or was overexpressed by recombinant CCL2 lentivirus, and cell viability was determined. An MTT assay indicated that CCL2 downregulation decreased the viability of A549 cells and augmented the DTX-induced cytotoxicity, whereas CCL2 upregulation protected A549 cells from DTX-induced cytotoxicity. Additionally, it was revealed that CCL2 overexpression activated phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling and inhibited apoptosis-associated protein caspase-3 activation and B-cell lymphoma 2 (Bcl-2) phosphorylation at Ser70 induced by DTX, and enhanced DTX-induced Bcl-2-associated death promoter phosphorylation at Ser112. PI3K/AKT inhibitor LY294002 restored DTX-induced caspase-3 activation and Bcl-2 phosphorylation, reversed the effect of CCL2 on the viability of A549 cells and enhanced DTX-induced cytotoxicity. These results demonstrated that chemoresistance may be mediated by cell stress responses involving CCL2 expression, suggesting that CCL2 may be a potential target for enhancing the therapeutic effect of DTX in lung cancer.
Collapse
Affiliation(s)
- Ting Wang
- Department of Oncology, The People's Hospital of Nanchang County, Nanchang, Jiangxi 330200, P.R. China
| | - Qingyuan Zhan
- Department of Internal Medicine 2, Tumor Hospital of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaodong Peng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhimin Qiu
- Department of Internal Medicine 6, Tumor Hospital of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Tiantian Zhao
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
12
|
Wilson KR, Kang IH, Baliga U, Xiong Y, Chatterjee S, Moore E, Parthiban B, Thyagarajan K, Borke JL, Mehrotra S, Kirkwood KL, LaRue AC, Ogawa M, Mehrotra M. Hematopoietic Stem Cells as a Novel Source of Dental Tissue Cells. Sci Rep 2018; 8:8026. [PMID: 29795229 PMCID: PMC5966408 DOI: 10.1038/s41598-018-26258-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 05/08/2018] [Indexed: 12/19/2022] Open
Abstract
While earlier studies have suggested that cells positive for hematopoietic markers can be found in dental tissues, it has yet to be confirmed. To conclusively demonstrate this, we utilized a unique transgenic model in which all hematopoietic cells are green fluorescent protein+ (GFP+). Pulp, periodontal ligament (PDL) and alveolar bone (AvB) cell culture analysis demonstrated numerous GFP+ cells, which were also CD45+ (indicating hematopoietic origin) and co-expressed markers of cellular populations in pulp (dentin matrix protein-1, dentin sialophosphoprotein, alpha smooth muscle actin [ASMA], osteocalcin), in PDL (periostin, ASMA, vimentin, osteocalcin) and in AvB (Runx-2, bone sialoprotein, alkaline phosphatase, osteocalcin). Transplantation of clonal population derived from a single GFP+ hematopoietic stem cell (HSC), into lethally irradiated recipient mice, demonstrated numerous GFP+ cells within dental tissues of recipient mice, which also stained for markers of cell populations in pulp, PDL and AvB (used above), indicating that transplanted HSCs can differentiate into cells in dental tissues. These hematopoietic-derived cells deposited collagen and can differentiate in osteogenic media, indicating that they are functional. Thus, our studies demonstrate, for the first time, that cells in pulp, PDL and AvB can have a hematopoietic origin, thereby opening new avenues of therapy for dental diseases and injuries.
Collapse
Affiliation(s)
- Katie R Wilson
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - In-Hong Kang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Uday Baliga
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Ying Xiong
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Shilpak Chatterjee
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Emily Moore
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Beneta Parthiban
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | | | - James L Borke
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Keith L Kirkwood
- Department of Oral Biology, University at Buffalo, The State University of New York, Department of Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14260, USA
| | - Amanda C LaRue
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.,Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA.,Ralph H Johnson VA Medical Center, Charleston, SC, 29425, USA
| | - Makio Ogawa
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Meenal Mehrotra
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA. .,Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA. .,Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA. .,Center for Oral Health Research, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
13
|
Liu FD, Pishesha N, Poon Z, Kaushik T, Van Vliet KJ. Material Viscoelastic Properties Modulate the Mesenchymal Stem Cell Secretome for Applications in Hematopoietic Recovery. ACS Biomater Sci Eng 2017; 3:3292-3306. [DOI: 10.1021/acsbiomaterials.7b00644] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Frances D. Liu
- Department
of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- BioSystems
and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore−MIT Alliance for Research and Technology, CREATE, Singapore 138602
| | - Novalia Pishesha
- Department
of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Zhiyong Poon
- BioSystems
and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore−MIT Alliance for Research and Technology, CREATE, Singapore 138602
| | - Tanwi Kaushik
- BioSystems
and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore−MIT Alliance for Research and Technology, CREATE, Singapore 138602
| | - Krystyn J. Van Vliet
- Department
of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- BioSystems
and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore−MIT Alliance for Research and Technology, CREATE, Singapore 138602
- Department
of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
14
|
McDonald LT, Johnson SD, Russell DL, Young MRI, LaRue AC. Role of a novel immune modulating DDR2-expressing population in silica-induced pulmonary fibrosis. PLoS One 2017; 12:e0180724. [PMID: 28700752 PMCID: PMC5507261 DOI: 10.1371/journal.pone.0180724] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 06/20/2017] [Indexed: 12/31/2022] Open
Abstract
Micro-injuries associated with chronic inhaled particle exposures are linked with activation of the immune response and are thought to contribute to progression of fibrotic disease. In the pulmonary environment, we have previously demonstrated a heterogeneous population of circulating fibroblast precursors (CFPs), which are defined by expression of the pan-leukocyte marker CD45 and the collagen receptor, discoidin domain receptor-2 (DDR2). This population is derived from the hematopoietic stem cell, expresses collagen, and has a fibroblastic morphology in vitro. Herein, we demonstrate a novel subset of CFPs expressing immune markers CD11b, CD11c, and major histocompatibility complex II (MHC II). The CFP population was skewed toward this immune marker expressing subset in animals with silica-induced pulmonary fibrosis. Data indicate that this CFP subset upregulates co-stimulatory molecules and MHC II expression in response to silica-induced fibrosis in vivo. Functionally, this population was shown to promote T cell skewing away from a Th1 response and toward a pro-inflammatory profile. These studies represent the first direct flow cytometric and functional evaluation of the novel immune marker expressing CFP subset in an exposure-induced model of pulmonary fibrosis. Elucidating the role of this CFP subset may enhance our understanding of the complex immune balance critical to mediating exposures at the pulmonary-host interface and may be a valuable target for the treatment of exposure-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Lindsay T. McDonald
- Research Services, Ralph H. Johnson VA Medical Center, Charleston, South Carolina, United States of America
- The Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Sara D. Johnson
- Research Services, Ralph H. Johnson VA Medical Center, Charleston, South Carolina, United States of America
- The Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Dayvia L. Russell
- Research Services, Ralph H. Johnson VA Medical Center, Charleston, South Carolina, United States of America
- The Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - M. Rita I. Young
- Research Services, Ralph H. Johnson VA Medical Center, Charleston, South Carolina, United States of America
- The Department of Otolaryngology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Amanda C. LaRue
- Research Services, Ralph H. Johnson VA Medical Center, Charleston, South Carolina, United States of America
- The Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
15
|
Fujita S, Ikeda T. The CCL2-CCR2 Axis in Lymph Node Metastasis From Oral Squamous Cell Carcinoma: An Immunohistochemical Study. J Oral Maxillofac Surg 2017; 75:742-749. [DOI: 10.1016/j.joms.2016.09.052] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/28/2016] [Accepted: 09/28/2016] [Indexed: 12/13/2022]
|
16
|
Hematopoietic stem cell-derived cancer-associated fibroblasts are novel contributors to the pro-tumorigenic microenvironment. Neoplasia 2016; 17:434-48. [PMID: 26025666 PMCID: PMC4468366 DOI: 10.1016/j.neo.2015.04.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 04/10/2015] [Accepted: 04/24/2015] [Indexed: 02/07/2023] Open
Abstract
Targeting the tumor microenvironment is critical toward improving the effectiveness of cancer therapeutics. Cancer-associated fibroblasts (CAFs) are one of the most abundant cell types of the tumor microenvironment, playing an important role in tumor progression. Multiple origins for CAFs have been proposed including resident fibroblasts, adipocytes, and bone marrow. Our laboratory previously identified a novel hematopoietic stem cell (HSC) origin for CAFs; however, the functional roles of HSC-derived CAFs (HSC-CAFs) in tumor progression have not yet been examined. To test the hypothesis that HSC-CAFs promote tumor progression through contribution to extracellular matrix (ECM) and paracrine production of pro-angiogenic factors, we developed a method to isolate HSC-CAFs. HSC-CAFs were profiled on the basis of their expression of hematopoietic and fibroblastic markers in two murine tumor models. Profiling revealed production of factors associated with ECM deposition and remodeling. Functional in vivo studies showed that co-injection of HSC-CAFs with tumor cells resulted in increased tumor growth rate and significantly larger tumors than tumor cells alone. Immunohistochemical studies revealed increased blood vessel density with co-injection, demonstrating a role for HSC-CAFs in tumor vascularization. Mechanistic in vitro studies indicated that HSC-CAFs play a role in producing vascular endothelial growth factor A and transforming growth factor–β1 in endothelial tube formation and patterning. In vitro and in vivo findings suggest that HSC-CAFs are a critical component of the tumor microenvironment and suggest that targeting the novel HSC-CAF may be a promising therapeutic strategy.
Collapse
|
17
|
Abstract
The understanding of bone marrow stem cell plasticity and contribution of bone marrow stem cells to pathophysiology is evolving with the advent of innovative technologies. Recent data has led to new mechanistic insights in the field of mesenchymal stem cell (MSC) research, and an increased appreciation for the plasticity of the hematopoietic stem cell (HSC). In this review, we discuss current research examining the origin of pulmonary cell types from endogenous lung stem and progenitor cells as well as bone marrow-derived stem cells (MSCs and HSCs) and their contributions to lung homeostasis and pathology. We specifically highlight recent findings from our laboratory that demonstrate an HSC origin for pulmonary fibroblasts based on transplantation of a clonal population of cells derived from a single HSC. These findings demonstrate the importance of developing an understanding of the sources of effector cells in disease state. Finally, a perspective is given on the potential clinical implications of these studies and others addressing stem cell contributions to lung tissue homeostasis and pathology.
Collapse
Affiliation(s)
- Lindsay T McDonald
- Research Services, Ralph H Johnson VAMC, Charleston, SC, USA; Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Amanda C LaRue
- Research Services, Ralph H Johnson VAMC, Charleston, SC, USA; Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
18
|
Hematopoietic Origin of Murine Lung Fibroblasts. Stem Cells Int 2015; 2015:159713. [PMID: 26185498 PMCID: PMC4491389 DOI: 10.1155/2015/159713] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 05/21/2015] [Accepted: 05/25/2015] [Indexed: 12/14/2022] Open
Abstract
Multiple origins, including the bone marrow, have been suggested to contribute to fibroblast populations in the lung. Using bone marrow reconstitution strategies, the present study tested the hypothesis that the bone marrow hematopoietic stem cell (HSC) gives rise to lung tissue fibroblasts in vivo. Data demonstrate that the nonadherent bone marrow fraction is enriched for CD45+ HSC-derived cells and was able to reconstitute hematopoiesis in lethally irradiated animals. Analysis of peripheral blood and lung tissues from engrafted mice demonstrated the ability of this population to give rise to CD45+/Discoidin-Domain Receptor-2+ (DDR2) circulating fibroblast precursors (CFPs) in blood and fibroblast populations in lung. An HSC origin for lung fibroblasts was confirmed using a novel clonal cell transplantation method in which the bone marrow is reconstituted by a clonal population derived from a single HSC. Together, these findings provide evidence for an HSC contribution to lung fibroblasts and demonstrate a circulating intermediate through the CD45+/DDR2+ HSC-derived CFP.
Collapse
|
19
|
Xiong Y, McDonald LT, Russell DL, Kelly RR, Wilson KR, Mehrotra M, Soloff AC, LaRue AC. Hematopoietic stem cell-derived adipocytes and fibroblasts in the tumor microenvironment. World J Stem Cells 2015; 7:253-265. [PMID: 25815113 PMCID: PMC4369485 DOI: 10.4252/wjsc.v7.i2.253] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 11/08/2014] [Accepted: 12/01/2014] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment (TME) is complex and constantly evolving. This is due, in part, to the crosstalk between tumor cells and the multiple cell types that comprise the TME, which results in a heterogeneous population of tumor cells and TME cells. This review will focus on two stromal cell types, the cancer-associated adipocyte (CAA) and the cancer-associated fibroblast (CAF). In the clinic, the presence of CAAs and CAFs in the TME translates to poor prognosis in multiple tumor types. CAAs and CAFs have an activated phenotype and produce growth factors, inflammatory factors, cytokines, chemokines, extracellular matrix components, and proteases in an accelerated and aberrant fashion. Through this activated state, CAAs and CAFs remodel the TME, thereby driving all aspects of tumor progression, including tumor growth and survival, chemoresistance, tumor vascularization, tumor invasion, and tumor cell metastasis. Similarities in the tumor-promoting functions of CAAs and CAFs suggest that a multipronged therapeutic approach may be necessary to achieve maximal impact on disease. While CAAs and CAFs are thought to arise from tissues adjacent to the tumor, multiple alternative origins for CAAs and CAFs have recently been identified. Recent studies from our lab and others suggest that the hematopoietic stem cell, through the myeloid lineage, may serve as a progenitor for CAAs and CAFs. We hypothesize that the multiple origins of CAAs and CAFs may contribute to the heterogeneity seen in the TME. Thus, a better understanding of the origin of CAAs and CAFs, how this origin impacts their functions in the TME, and the temporal participation of uniquely originating TME cells may lead to novel or improved anti-tumor therapeutics.
Collapse
|
20
|
Tang X, Amar S. p53 suppresses CCL2-induced subcutaneous tumor xenograft. Tumour Biol 2014; 36:2801-8. [PMID: 25492482 DOI: 10.1007/s13277-014-2906-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 11/27/2014] [Indexed: 11/30/2022] Open
Abstract
Chemokine (C-C motif) ligand 2 (CCL2) has recently been found to be a key player in the pathology of many human glomerular and tubulointerstitial diseases. CCL2 has also been found to be expressed in various cancers, including human hepatoma cells, human cancer progression, and human multiple myeloma cells. Thus, the inhibition of elevated CCL2 production may provide a new avenue for therapeutic intervention in CCL2-mediated cancer diseases. A previous study has indicated that knockdown of human p53 has a strong negative impact on CCL2 induction. We therefore are interested in how p53 regulates CCL2 gene expression. In the following study, our findings indicate that p53 binds to CCL2, consequently significantly downregulating CCL2 promoter activity. Furthermore, injection of CCL2-promoting cancer cells (CCL2/A549) in p53-deficient mice for 3 weeks strongly induced subcutaneous xenograft tumor growth compared with the control. Overall, the research results support the novel role of p53 in suppression of chemokine (such as CCL2)-mediated cancer diseases.
Collapse
Affiliation(s)
- Xiaoren Tang
- Center for Anti-Inflammatory Therapeutics, Department of Molecular & Cell Biology, Boston University Goldman School of Dental Medicine, 650 Albany Street, X-343, Boston, MA, 02118, USA
| | | |
Collapse
|
21
|
Mamlouk S, Kalucka J, Singh RP, Franke K, Muschter A, Langer A, Jakob C, Gassmann M, Baretton GB, Wielockx B. Loss of prolyl hydroxylase-2 in myeloid cells and T-lymphocytes impairs tumor development. Int J Cancer 2013; 134:849-58. [DOI: 10.1002/ijc.28409] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 06/14/2013] [Accepted: 07/16/2013] [Indexed: 12/20/2022]
Affiliation(s)
- Soulafa Mamlouk
- Emmy Noether Research Group; University of Technology; Dresden Germany
- Institute of Pathology; University of Technology Dresden; Dresden Germany
| | - Joanna Kalucka
- Emmy Noether Research Group; University of Technology; Dresden Germany
- Institute of Pathology; University of Technology Dresden; Dresden Germany
| | - Rashim Pal Singh
- Emmy Noether Research Group; University of Technology; Dresden Germany
- Institute of Pathology; University of Technology Dresden; Dresden Germany
| | - Kristin Franke
- Emmy Noether Research Group; University of Technology; Dresden Germany
- Institute of Pathology; University of Technology Dresden; Dresden Germany
| | - Antje Muschter
- Emmy Noether Research Group; University of Technology; Dresden Germany
- Institute of Pathology; University of Technology Dresden; Dresden Germany
| | - Anika Langer
- Emmy Noether Research Group; University of Technology; Dresden Germany
- Institute of Pathology; University of Technology Dresden; Dresden Germany
| | - Christiane Jakob
- Institute of Pathology; University of Technology Dresden; Dresden Germany
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty and Zurich Center for Integrative Human Physiology (ZIHP); University of Zürich; Zürich Switzerland
- Universidad Peruana Cayetano Heredia (UPCH); Lima Peru
| | | | - Ben Wielockx
- Emmy Noether Research Group; University of Technology; Dresden Germany
- Institute of Pathology; University of Technology Dresden; Dresden Germany
- DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden; University of Technology Dresden; Dresden Germany
| |
Collapse
|
22
|
Raynaud CM, Butler JM, Halabi NM, Ahmad FS, Ahmed B, Rafii S, Rafii A. Endothelial cells provide a niche for placental hematopoietic stem/progenitor cell expansion through broad transcriptomic modification. Stem Cell Res 2013; 11:1074-90. [PMID: 23978474 DOI: 10.1016/j.scr.2013.07.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 07/17/2013] [Accepted: 07/31/2013] [Indexed: 11/26/2022] Open
Abstract
Umbilical cord blood (UCB) is an attractive source of hematopoietic stem cells (HSCs). However, the number of HSCs in UCB is limited, and attempts to amplify them in vitro remain inefficient. Several publications have documented amplification of hematopoietic stem/progenitor cells (HSPCs) on endothelial or mesenchymal cells, but the lack of homogeneity in culture conditions and HSC definition impairs direct comparison of these results. We investigated the ability of different feeder layers, mesenchymal progenitors (MPs) and endothelial cells (ECs), to amplify hematopoietic stem/progenitor cells. Placental derived HSPCs (defined as Lin(-)CD45(-/dim)CD34(+)CD38(-)CD90(+)) were maintained on confluent feeder layers and the number of cells and their marker expression were monitored over 21 days. Although both types of feeder layers supported hematopoietic expansion, only endothelial cells triggered amplification of Lin(-)CD45(-/dim)CD34(+)CD38(-)CD90(+) cells, which peaked at 14 days. The amplified cells differentiated into all cell lineages, as attested by in vitro colony-forming assays, and were capable of engraftment and multi-lineage differentiation in sub-lethally irradiated mice. Mesenchymal progenitors promoted amplification of CD38(+) cells, previously defined as precursors with more limited differentiation potential. A competitive assay demonstrated that hematopoietic stem/progenitor cells had a preference for interacting with endothelial cells in vitro. Cytokine and transcriptomic analysis of both feeder cell types identified differences in gene expression that correlated with propensity of ECs and MPs to support hematopoietic cell amplification and differentiation respectively. Finally, we used RNA sequencing of endothelial cells and HSPCs to uncover relevant networks illustrating the complex interaction between endothelial cells and HSPCs leading to stem/progenitor cell expansion.
Collapse
Affiliation(s)
- Christophe M Raynaud
- Qatar Cardiovascular Research Center, Qatar Foundation, Qatar Science and Technology Park, Doha, Qatar
| | | | | | | | | | | | | |
Collapse
|
23
|
Tang X, Asano M, O'Reilly A, Farquhar A, Yang Y, Amar S. p53 is an important regulator of CCL2 gene expression. Curr Mol Med 2013; 12:929-43. [PMID: 22804246 DOI: 10.2174/156652412802480844] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 02/20/2012] [Accepted: 02/21/2012] [Indexed: 12/11/2022]
Abstract
The p53 protein is a sequence-specific DNA-binding factor that regulates inflammatory genes such as CCL2/MCP-1 that may play a role in various diseases. A recent study has indicated that the knockdown of human p53 leads to a strong negative regulation of CCL2 induction. We are therefore interested in how p53 regulates CCL2 gene expression. In the following study, our findings indicate that UV-induced p53 accumulation in mouse macrophages significantly decreases LPS-induced CCL2 production, and that p53 binds to CCL2 5'UTR in the region (16-35). We also found that a p53 domain (p53pep170) mimics full length p53 to down-regulate CCL2 promoter activity. Treatment of p53-deficient mouse primary macrophages with synthetic p53pep170 was found to decrease LPS-induced production of CCL2 without association with cellular endogenous p53. CCL2 production induced by lentiCLG in human monocytes or mouse primary macrophages was blocked in the presence of p53pep170. Overall, these results demonstrate that p53 or its derived peptide (p53pep170) is an important regulator of CCL2 gene expression via its binding activity, and acts as a novel model for future studies linking p53 and its short peptide to pave the way to possible pharmaceutical intervention of CCL2-mediated inflammatory and cancer diseases.
Collapse
Affiliation(s)
- X Tang
- Center for Anti- Inflammatory Therapeutics, Boston University, 650 Albany Street, X- 343, Boston, MA 02118, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Liu Y, Chen K, Wang C, Gong W, Yoshimura T, Liu M, Wang JM. Cell surface receptor FPR2 promotes antitumor host defense by limiting M2 polarization of macrophages. Cancer Res 2013; 73:550-60. [PMID: 23139214 PMCID: PMC3549056 DOI: 10.1158/0008-5472.can-12-2290] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
FPR2 (Fpr2 in mouse) is a G-protein-coupled receptor interacting with bacterial and host-derived chemotactic agonists. Fpr2 supports innate and adaptive immune responses as illustrated by the reduction in severity of allergic airway inflammation in Fpr2-KO mice, due to impaired trafficking of antigen-presenting dendritic cells (DC). The aim of this study is to examine the role of Fpr2 in host antitumor responses. We found that Fpr2-KO mice bearing subcutaneously implanted Lewis lung carcinoma (LLC) cells exhibited significantly shortened survival than normal mice due to more rapidly growing tumors. In contrast, in Fpr2-transgenic mice overexpressing Fpr2, subcutaneously implanted LLC tumors grew more slowly than those in wild-type (WT) littermates. Investigation of tumor tissues revealed an increased number of macrophages associated with tumors grown in Fpr2-KO mice. Macrophages derived from Fpr2-KO mice showed a more potent chemotactic response to LLC-derived supernatant (LLC Sup), which could be neutralized by an anti-CCL2 antibody. The increased chemotaxis of Fpr2-KO mouse macrophages in response to LLC Sup was due to their higher level expression of CCR4, a chemokine receptor that also recognizes the ligand CCL2. Furthermore, macrophages from Fpr2-KO mice acquired an M2 phenotype after stimulation with LLC Sup. These results suggest that Fpr2 plays an important role in host defense against implanted LLC by sustaining macrophages in an M1 phenotype with more potent antitumor activities.
Collapse
Affiliation(s)
- Ying Liu
- Laboratory of Molecular Immunoregulation, Cancer Inflammation Program, Center of Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Maryland, 21702, USA
| | - Keqiang Chen
- Laboratory of Molecular Immunoregulation, Cancer Inflammation Program, Center of Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Maryland, 21702, USA
| | - Chunyan Wang
- Laboratory of Molecular Immunoregulation, Cancer Inflammation Program, Center of Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Maryland, 21702, USA
- Xuzhou Yes Biotech Laboratories Ltd. Xuzhou, Jiangsu, 221004, China
| | - Wanghua Gong
- Basic Research Program, SAIC-Frederick, Frederick, Maryland, 21702, USA
| | - Teizo Yoshimura
- Laboratory of Molecular Immunoregulation, Cancer Inflammation Program, Center of Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Maryland, 21702, USA
| | - Mingyong Liu
- Laboratory of Molecular Immunoregulation, Cancer Inflammation Program, Center of Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Maryland, 21702, USA
- Department of Spine Surgery, Daping Hospital, Third Military Medical University, Chongqing, 400042, P. R. China
| | - Ji Ming Wang
- Laboratory of Molecular Immunoregulation, Cancer Inflammation Program, Center of Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Maryland, 21702, USA
| |
Collapse
|
25
|
Lin S, Fu S. The correlation of hematopoietic stem cells with cancer stem cells through the regulation of stromal cells in tumor microenvironment. Med Hypotheses 2013; 80:494-7. [PMID: 23317540 DOI: 10.1016/j.mehy.2012.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 12/09/2012] [Accepted: 12/13/2012] [Indexed: 01/15/2023]
Abstract
Cancer stem cells are a small population of tumor cells that have many malignant features such as chemotherapy resistance, radiotherapy resistance, tumorigenicity and are responsible for tumor progression, disease recurrence and metastasis. Therefore, insight into the regulation of the biology of cancer stem cells is important to eradicate cancer. Recently, studies suggested that hematopoietic stem cells could incorporate into tumor stroma and differentiated into stromal cells and the cells derived from hematopoietic stem cells play an important role on tumor progress. Moreover, cancer cells competed with hematopoietic stem cells for occupancy of the hematopoietic stem cell niches to regulate bone metastasis and most cancer cells in bone marrow metastasis were cancer stem cells. Therefore, we hypothesize that cancer stem cells could promote hematopoietic stem cells incorporating into tumor microenvironment and resulting into transformation of hematopoietic stem cells to stromal cells, which could impact the biological behavior of cancer stem cells.
Collapse
Affiliation(s)
- Shuchen Lin
- Department of Radiation Oncology, Sixth People's Hospital of Jiao Tong University, Shanghai, People's Republic of China
| | | |
Collapse
|
26
|
McDonald LT, LaRue AC. Hematopoietic stem cell derived carcinoma-associated fibroblasts: a novel origin. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2012; 5:863-873. [PMID: 23119103 PMCID: PMC3484486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 09/23/2012] [Indexed: 06/01/2023]
Abstract
Researchers have begun to appreciate the significant role that the microenvironment plays in tumorigenesis and are now shedding light on the role of the stroma in induction and progression of solid tumors. While the stroma of solid tumors is comprised of many cell types, including vascular and immune cells, one of the most prominent cell types in the tumor stroma is the fibroblast, called the carcinoma-associated fibroblast (CAF) or tumor-associated fibroblast (TAF). The interaction between CAFs and tumor cells is quite complex. CAFs have been implicated in tumor angiogenesis, immunosuppression, tumor cell proliferation and aggressiveness, genetic instability, and metastasis. However, their specific roles in each of these processes have only been partially elucidated. Determining the role of CAFs has been complicated by the fact that researchers have demonstrated heterogeneity in the stromal fibroblast population. This heterogeneity has brought about the concept of multiple origins for CAFs. While many origins of CAFs have been suggested, in our own laboratory we have identified a novel hematopoietic stem cell (HSC) origin of CAFs. Given the profound role of CAFs in tumor progression and prognosis, the CAF represents an exciting potential therapeutic target. The heterogeneity of the CAF population makes research directed at investigating the roles and origins of CAFs critical to development of such anti-tumor therapies.
Collapse
Affiliation(s)
- Lindsay T McDonald
- The Department of Pathology and Laboratory Medicine, Medical University of South CarolinaSC
- Hollings Cancer Center, Medical University of South CarolinaSC
- Research Services, Ralph H. Johnson Veterans Affairs Medical CenterCharleston, SC
| | - Amanda C LaRue
- The Department of Pathology and Laboratory Medicine, Medical University of South CarolinaSC
- Hollings Cancer Center, Medical University of South CarolinaSC
- Research Services, Ralph H. Johnson Veterans Affairs Medical CenterCharleston, SC
| |
Collapse
|
27
|
Mehrotra M, Williams CR, Ogawa M, LaRue AC. Hematopoietic stem cells give rise to osteo-chondrogenic cells. Blood Cells Mol Dis 2012; 50:41-9. [PMID: 22954476 DOI: 10.1016/j.bcmd.2012.08.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2012] [Revised: 08/08/2012] [Accepted: 08/08/2012] [Indexed: 12/15/2022]
Abstract
Repair of bone fracture requires recruitment and proliferation of stem cells with the capacity to differentiate to functional osteoblasts. Given the close association of bone and bone marrow (BM), it has been suggested that BM may serve as a source of these progenitors. To test the ability of hematopoietic stem cells (HSCs) to give rise to osteo-chondrogenic cells, we used a single HSC transplantation paradigm in uninjured bone and in conjunction with a tibial fracture model. Mice were lethally irradiated and transplanted with a clonal population of cells derived from a single enhanced green fluorescent protein positive (eGFP+) HSC. Analysis of paraffin sections from these animals showed the presence of eGFP+ osteocytes and hypertrophic chondrocytes. To determine the contribution of HSC-derived cells to fracture repair, non-stabilized tibial fracture was created. Paraffin sections were examined at 7 days, 2 weeks and 2 months after fracture and eGFP+ hypertrophic chondrocytes, osteoblasts and osteocytes were identified at the callus site. These cells stained positive for Runx-2 or osteocalcin and also stained for eGFP demonstrating their origin from the HSC. Together, these findings strongly support the concept that HSCs generate bone cells and suggest therapeutic potentials of HSCs in fracture repair.
Collapse
Affiliation(s)
- Meenal Mehrotra
- Department of Veterans Affairs Medical Center, Ralph H. Johnson VAMC, Medical University of South Carolina, Charleston, SC, USA
| | | | | | | |
Collapse
|