1
|
Petinati NA, Sadovskaya AV, Sats NV, Kapranov NM, Davydova YO, Fastova EA, Magomedova AU, Vasilyeva AN, Aleshina OA, Arapidi GP, Shender VO, Smirnov IP, Pobeguts OV, Lagarkova MA, Drize NI, Parovichnikova EN. Molecular Changes in Immunological Characteristics of Bone Marrow Multipotent Mesenchymal Stromal Cells in Lymphoid Neoplasia. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:883-903. [PMID: 38880649 DOI: 10.1134/s0006297924050092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 06/18/2024]
Abstract
Immune system and bone marrow stromal cells play an important role in maintaining normal hematopoiesis. Lymphoid neoplasia disturbs not only development of immune cells, but other immune response mechanisms as well. Multipotent mesenchymal stromal cells (MSCs) of the bone marrow are involved in immune response regulation through both intercellular interactions and secretion of various cytokines. In hematological malignancies, the bone marrow stromal microenvironment, including MSCs, is altered. Aim of this study was to describe the differences of MSCs' immunological function in the patients with acute lymphoblastic leukemia (ALL) and diffuse large B-cell lymphoma (DLBCL). In ALL, malignant cells arise from the early precursor cells localized in bone marrow, while in DLBCL they arise from more differentiated B-cells. In this study, only the DLBCL patients without bone marrow involvement were included. Growth parameters, surface marker expression, genes of interest expression, and secretion pattern of bone marrow MSCs from the patients with ALL and DLBCL at the onset of the disease and in remission were studied. MSCs from the healthy donors of corresponding ages were used as controls. It has been shown that concentration of MSCs in the bone marrow of the patients with ALL is reduced at the onset of the disease and is restored upon reaching remission; in the patients with DLBCL this parameter does not change. Proliferative capacity of MSCs did not change in the patients with ALL; however, the cells of the DLBCL patients both at the onset and in remission proliferated significantly faster than those from the donors. Expression of the membrane surface markers and expression of the genes important for differentiation, immunological status maintenance, and cytokine secretion differed significantly in the MSCs of the patients from those of the healthy donors and depended on nosology of the disease. Secretomes of the MSCs varied greatly; a number of proteins associated with immune response regulation, differentiation, and maintenance of hematopoietic stem cells were depleted in the secretomes of the cells from the patients. Lymphoid neoplasia leads to dramatic changes in the functional immunological status of MSCs.
Collapse
Affiliation(s)
- Nataliya A Petinati
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia.
| | - Aleksandra V Sadovskaya
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
- Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Natalia V Sats
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Nikolai M Kapranov
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Yulia O Davydova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Ekaterina A Fastova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Aminat U Magomedova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Anastasia N Vasilyeva
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Olga A Aleshina
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Georgiy P Arapidi
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, 141700, Russia
| | - Viktoria O Shender
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Igor P Smirnov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Olga V Pobeguts
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Maria A Lagarkova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Nina I Drize
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Elena N Parovichnikova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| |
Collapse
|
2
|
Li J, Li XL, Li CQ. Immunoregulation mechanism of VEGF signaling pathway inhibitors and its efficacy on the kidney. Am J Med Sci 2023; 366:404-412. [PMID: 37699444 DOI: 10.1016/j.amjms.2023.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 05/10/2023] [Accepted: 09/07/2023] [Indexed: 09/14/2023]
Abstract
Angiogenesis and immunosuppression are closely related pathophysiologic processes. Widely prescribed in malignant tumor and proliferative retinal lesions, VEGF signaling pathway inhibitors may cause hypertension and renal injury in some patients, presenting with proteinuria, nephrotic syndrome, renal failure and thrombotic microangiopathy. VEGF signaling pathway inhibitors block the action of both VEGF-A and VEGF-C. However, VEGF-A and VEGF-C produced by podocytes are vital to maintain the physiological function of glomerular endothelial cells and podocytes. There is still no effective treatment for kidney disease associated with VEGF signaling pathway inhibitors and some patients have progressive renal failure even after withdrawal of the drug. Recent studies reveal that blocking of VEGF-A and VEGF-C can activate CD4 +and CD8+ T cells, augment antigen-presenting function of dendritic cells, enhance cytotoxicity of macrophages and initiate complement cascade activation. VEGF and VEGFR are expressed in immune cells, which are involved in the immunosuppression and cross-talk among immune cells. This review summarizes the expression and function of VEGF-A and VEGF-C in the kidney. The current immunoregulation mechanisms of VEGF signaling pathway inhibitors are reviewed. Finally, combinate strategies are summarized to highlight the proposal for VEGF signaling pathway inhibitors.
Collapse
Affiliation(s)
- Jun Li
- Department of Nephrology, Affiliated Hospital of Jiangnan University, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Jiangsu, China.
| | - Xiao-Lin Li
- Wuxi School of Medicine, Jiangnan University, Jiangsu, China
| | - Chun-Qing Li
- Department of Nephrology, Affiliated Hospital of Jiangnan University, Jiangsu, China
| |
Collapse
|
3
|
Bone Marrow Stromal Cell Regeneration Profile in Treated B-Cell Precursor Acute Lymphoblastic Leukemia Patients: Association with MRD Status and Patient Outcome. Cancers (Basel) 2022; 14:cancers14133088. [PMID: 35804860 PMCID: PMC9265080 DOI: 10.3390/cancers14133088] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 12/10/2022] Open
Abstract
For the last two decades, measurable residual disease (MRD) has become one of the most powerful independent prognostic factors in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). However, the effect of therapy on the bone marrow (BM) microenvironment and its potential relationship with the MRD status and disease free survival (DFS) still remain to be investigated. Here we analyzed the distribution of mesenchymal stem cells (MSC) and endothelial cells (EC) in the BM of treated BCP-ALL patients, and its relationship with the BM MRD status and patient outcome. For this purpose, the BM MRD status and EC/MSC regeneration profile were analyzed by multiparameter flow cytometry (MFC) in 16 control BM (10 children; 6 adults) and 1204 BM samples from 347 children and 100 adult BCP-ALL patients studied at diagnosis (129 children; 100 adults) and follow-up (824 childhood samples; 151 adult samples). Patients were grouped into a discovery cohort (116 pediatric BCP-ALL patients; 338 samples) and two validation cohorts (74 pediatric BCP-ALL, 211 samples; and 74 adult BCP-ALL patients; 134 samples). Stromal cells (i.e., EC and MSC) were detected at relatively low frequencies in all control BM (16/16; 100%) and in most BCP-ALL follow-up samples (874/975; 90%), while they were undetected in BCP-ALL BM at diagnosis. In control BM samples, the overall percentage of EC plus MSC was higher in children than adults (p = 0.011), but with a similar EC/MSC ratio in both groups. According to the MRD status similar frequencies of both types of BM stromal cells were detected in BCP-ALL BM studied at different time points during the follow-up. Univariate analysis (including all relevant prognostic factors together with the percentage of stromal cells) performed in the discovery cohort was used to select covariates for a multivariate Cox regression model for predicting patient DFS. Of note, an increased percentage of EC (>32%) within the BCP-ALL BM stromal cell compartment at day +78 of therapy emerged as an independent unfavorable prognostic factor for DFS in childhood BCP-ALL in the discovery cohort—hazard ratio (95% confidence interval) of 2.50 (1−9.66); p = 0.05—together with the BM MRD status (p = 0.031). Further investigation of the predictive value of the combination of these two variables (%EC within stromal cells and MRD status at day +78) allowed classification of BCP-ALL into three risk groups with median DFS of: 3.9, 3.1 and 1.1 years, respectively (p = 0.001). These results were confirmed in two validation cohorts of childhood BCP-ALL (n = 74) (p = 0.001) and adult BCP-ALL (n = 40) (p = 0.004) treated at different centers. In summary, our findings suggest that an imbalanced EC/MSC ratio in BM at day +78 of therapy is associated with a shorter DFS of BCP-ALL patients, independently of their MRD status. Further prospective studies are needed to better understand the pathogenic mechanisms involved.
Collapse
|
4
|
Meena R, Nangia A, Sharma S, Chandra J. Serum Levels of Vascular Endothelial Growth Factor and Its Receptor in Newly Diagnosed Paediatric Acute Lymphoblastic Leukemia. Indian J Hematol Blood Transfus 2021; 37:586-592. [PMID: 34744342 PMCID: PMC8523739 DOI: 10.1007/s12288-021-01413-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/09/2021] [Indexed: 10/22/2022] Open
Abstract
Autocrine and paracrine loop involving vascular endothelial growth factor (VEGF) and its receptor have been described in haematological malignancies. However, scarce literature is present on angiogenesis in paediatric acute lymphoblastic leukemia (ALL) with studies showing controversial results. The aim was to study serum levels of VEGF and its receptors in paediatric ALL at the time of diagnosis and at the end of induction phase and to compare these levels with clinico-haematological parameters in these patients. Serum VEGF, VEGFR-1 and VEGFR-2 levels were measured by enzyme-linked immunoabsorbant assay at diagnosis (day 0) and at the end of induction phase (day 35) in 30 newly diagnosed paediatric ALL patients and in 10 healthy controls. Median s-VEGF was significantly lower at day 0 as compared to day 35 (196.15 vs. 606.75 pg/ml: p < 0.001). s-VEGFR-1 levels were detectable only in 7 patients at day 0 and were below detection level at day 35 in all patients. Median s-VEGFR-2 at day 0 was significantly lower as compared to day 35 (17,577.5 vs. 20,507.5 pg/ml; p = 0.005). Median VEGF-R1 showed an inverse relationship with VEGF-R2 but was statistically insignificant. All patients were in remission at the end of induction. Thus, leukemic infiltration of bone marrow affects angiogenesis and reduces pro-angiogenic markers VEGF and VEGFR-2 in serum possibly due to increased local consumption by blasts. A successful induction leads to clearing of blasts causing restoration of normal hematopoiesis with normalization of VEGF and VEGFR-2 levels.
Collapse
Affiliation(s)
- Rachana Meena
- Department of Pathology, Lady Hardinge Medical College, New Delhi, India
| | - Anita Nangia
- Department of Pathology, Lady Hardinge Medical College, New Delhi, India
| | - Sunita Sharma
- Department of Pathology, Lady Hardinge Medical College, New Delhi, India
| | - Jagdish Chandra
- Department of Paediatrics, Lady Hardinge Medical College, New Delhi, India
| |
Collapse
|
5
|
Ceci C, Atzori MG, Lacal PM, Graziani G. Role of VEGFs/VEGFR-1 Signaling and its Inhibition in Modulating Tumor Invasion: Experimental Evidence in Different Metastatic Cancer Models. Int J Mol Sci 2020; 21:E1388. [PMID: 32085654 PMCID: PMC7073125 DOI: 10.3390/ijms21041388] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 12/14/2022] Open
Abstract
The vascular endothelial growth factor (VEGF) family members, VEGF-A, placenta growth factor (PlGF), and to a lesser extent VEGF-B, play an essential role in tumor-associated angiogenesis, tissue infiltration, and metastasis formation. Although VEGF-A can activate both VEGFR-1 and VEGFR-2 membrane receptors, PlGF and VEGF-B exclusively interact with VEGFR-1. Differently from VEGFR-2, which is involved both in physiological and pathological angiogenesis, in the adult VEGFR-1 is required only for pathological angiogenesis. Besides this role in tumor endothelium, ligand-mediated stimulation of VEGFR-1 expressed in tumor cells may directly induce cell chemotaxis and extracellular matrix invasion. Furthermore, VEGFR-1 activation in myeloid progenitors and tumor-associated macrophages favors cancer immune escape through the release of immunosuppressive cytokines. These properties have prompted a number of preclinical and clinical studies to analyze VEGFR-1 involvement in the metastatic process. The aim of the present review is to highlight the contribution of VEGFs/VEGFR-1 signaling in the progression of different tumor types and to provide an overview of the therapeutic approaches targeting VEGFR-1 currently under investigation.
Collapse
Affiliation(s)
- Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.C.); (M.G.A.)
| | - Maria Grazia Atzori
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.C.); (M.G.A.)
| | - Pedro Miguel Lacal
- Laboratory of Molecular Oncology, “Istituto Dermopatico dell’Immacolata-Istituto di Ricovero e Cura a Carattere Scientifico”, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy;
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.C.); (M.G.A.)
| |
Collapse
|
6
|
Central nervous system involvement in acute lymphoblastic leukemia is mediated by vascular endothelial growth factor. Blood 2017; 130:643-654. [PMID: 28550041 DOI: 10.1182/blood-2017-03-769315] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/21/2017] [Indexed: 12/12/2022] Open
Abstract
In acute lymphoblastic leukemia (ALL), central nervous system (CNS) involvement is a major clinical concern. Despite nondetectable CNS leukemia in many cases, prophylactic CNS-directed conventional intrathecal chemotherapy is required for relapse-free survival, indicating subclinical CNS manifestation in most patients. However, CNS-directed therapy is associated with long-term sequelae, including neurocognitive deficits and secondary neoplasms. Therefore, molecular mechanisms and pathways mediating leukemia-cell entry into the CNS need to be understood to identify targets for prophylactic and therapeutic interventions and develop alternative CNS-directed treatment strategies. In this study, we analyzed leukemia-cell entry into the CNS using a primograft ALL mouse model. We found that primary ALL cells transplanted onto nonobese diabetic/severe combined immunodeficiency mice faithfully recapitulated clinical and pathological features of meningeal infiltration seen in patients with ALL. ALL cells that had entered the CNS and were infiltrating the meninges were characterized by high expression of vascular endothelial growth factor A (VEGF). Although cellular viability, growth, proliferation, and survival of ALL cells were found to be independent of VEGF, transendothelial migration through CNS microvascular endothelial cells was regulated by VEGF. The importance of VEGF produced by ALL cells in mediating leukemia-cell entry into the CNS and leptomeningeal infiltration was further demonstrated by specific reduction of CNS leukemia on in vivo VEGF capture by the anti-VEGF antibody bevacizumab. Thus, we identified a mechanism of ALL-cell entry into the CNS, which by targeting VEGF signaling may serve as a novel strategy to control CNS leukemia in patients, replacing conventional CNS-toxic treatment.
Collapse
|
7
|
Wang W, Ma Y, Li J, Shi HS, Wang LQ, Guo FC, Zhang J, Li D, Mo BH, Wen F, Liu T, Liu YT, Wang YS, Wei YQ. Specificity redirection by CAR with human VEGFR-1 affinity endows T lymphocytes with tumor-killing ability and anti-angiogenic potency. Gene Ther 2013; 20:970-8. [PMID: 23636245 DOI: 10.1038/gt.2013.19] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 03/02/2013] [Accepted: 03/25/2013] [Indexed: 02/05/2023]
Abstract
Immunotherapy that is based on adoptive transfer of T lymphocytes, which are genetically modified to express chimeric antigen receptors (CARs) that recognize tumor-associated antigens, has been demonstrated to be an efficient cancer therapy. Vascular endothelial growth factor receptor-1 (VEGFR-1), a vital molecule involved in tumor growth and angiogenesis, has not been targeted by CAR-modified T lymphocytes. In this study, we generated CAR-modified T lymphocytes with human VEGFR-1 specificity (V-1 CAR) by electroporation. V-1 CAR-modified T lymphocytes were demonstrated to elicit lytic cytotoxicity to target cells in a VEGFR-1-dependent manner. The adoptive transfer of V-1 CAR T lymphocytes delayed tumor growth and formation and inhibited pulmonary metastasis in xenograft models and such efficacies were enhanced by cotransfer of T lymphocytes that expressed interleukin-15 (IL-15). Moreover, V-1 CAR-modified T lymphocytes lysed primary endothelial cells and impaired tube formation, in vitro. These data demonstrated the antitumor and anti-angiogenesis ability of V-1 CAR-modified T lymphocytes. Our study provides the rationale for the clinical translation of CAR-modified T lymphocytes with VEGFR-1 specificity.
Collapse
Affiliation(s)
- W Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Kottke T, Hall G, Pulido J, Diaz RM, Thompson J, Chong H, Selby P, Coffey M, Pandha H, Chester J, Melcher A, Harrington K, Vile R. Antiangiogenic cancer therapy combined with oncolytic virotherapy leads to regression of established tumors in mice. J Clin Invest 2010; 120:1551-60. [PMID: 20364090 PMCID: PMC2860921 DOI: 10.1172/jci41431] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2009] [Accepted: 01/27/2010] [Indexed: 12/31/2022] Open
Abstract
Clinical trials of oncolytic virotherapy have shown low toxicity and encouraging signs of efficacy. However, it remains critically important to develop methods for systemic viral delivery if such therapies are to be clinically implemented to treat established tumors. In this respect, much effort is being focused on combining oncolytic viruses with standard treatment modalities such as inhibitors of VEGF165 (an alternatively spliced isoform of VEGF-A) signaling, which are widely used to treat several different cancers. Here, we have demonstrated that combining VEGF165 inhibitors with systemic delivery of oncolytic viruses leads to substantial regression and cure of established tumors in immunocompetent mice. We have shown that manipulating VEGF165-mediated signaling by administering VEGF165 to mice harboring mouse melanoma cells that do not express VEGF165 and by administering a VEGF inhibitor and then withdrawing treatment to allow VEGF levels to rebound in mice harboring mouse melanoma cells expressing VEGF165 allows tumor-associated endothelial cells transiently to support viral replication. This approach led to direct tumor cell lysis and triggered innate immune-mediated attack on the tumor vasculature. It also resulted in long-term antitumor effects, even against tumors in which viral replication is poorly supported. Since this combinatorial approach targets the tumor endothelium, we believe these data have direct, wide-ranging, and immediate clinical applicability across a broad range of tumor types.
Collapse
Affiliation(s)
- Timothy Kottke
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Cancer Research UK Clinical Centre, St. James’ University Hospital, Leeds, United Kingdom.
Department of Ophthalmology and Ocular Oncology,
St George’s Hospital Medical School, Tooting, London, United Kingdom.
Oncolytics Biotech Inc., Calgary, Canada.
Department of Oncology, University of Surrey, Guildford, United Kingdom.
The Institute of Cancer Research, London, United Kingdom.
Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Geoff Hall
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Cancer Research UK Clinical Centre, St. James’ University Hospital, Leeds, United Kingdom.
Department of Ophthalmology and Ocular Oncology,
St George’s Hospital Medical School, Tooting, London, United Kingdom.
Oncolytics Biotech Inc., Calgary, Canada.
Department of Oncology, University of Surrey, Guildford, United Kingdom.
The Institute of Cancer Research, London, United Kingdom.
Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jose Pulido
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Cancer Research UK Clinical Centre, St. James’ University Hospital, Leeds, United Kingdom.
Department of Ophthalmology and Ocular Oncology,
St George’s Hospital Medical School, Tooting, London, United Kingdom.
Oncolytics Biotech Inc., Calgary, Canada.
Department of Oncology, University of Surrey, Guildford, United Kingdom.
The Institute of Cancer Research, London, United Kingdom.
Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Rosa Maria Diaz
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Cancer Research UK Clinical Centre, St. James’ University Hospital, Leeds, United Kingdom.
Department of Ophthalmology and Ocular Oncology,
St George’s Hospital Medical School, Tooting, London, United Kingdom.
Oncolytics Biotech Inc., Calgary, Canada.
Department of Oncology, University of Surrey, Guildford, United Kingdom.
The Institute of Cancer Research, London, United Kingdom.
Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jill Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Cancer Research UK Clinical Centre, St. James’ University Hospital, Leeds, United Kingdom.
Department of Ophthalmology and Ocular Oncology,
St George’s Hospital Medical School, Tooting, London, United Kingdom.
Oncolytics Biotech Inc., Calgary, Canada.
Department of Oncology, University of Surrey, Guildford, United Kingdom.
The Institute of Cancer Research, London, United Kingdom.
Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Heung Chong
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Cancer Research UK Clinical Centre, St. James’ University Hospital, Leeds, United Kingdom.
Department of Ophthalmology and Ocular Oncology,
St George’s Hospital Medical School, Tooting, London, United Kingdom.
Oncolytics Biotech Inc., Calgary, Canada.
Department of Oncology, University of Surrey, Guildford, United Kingdom.
The Institute of Cancer Research, London, United Kingdom.
Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Peter Selby
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Cancer Research UK Clinical Centre, St. James’ University Hospital, Leeds, United Kingdom.
Department of Ophthalmology and Ocular Oncology,
St George’s Hospital Medical School, Tooting, London, United Kingdom.
Oncolytics Biotech Inc., Calgary, Canada.
Department of Oncology, University of Surrey, Guildford, United Kingdom.
The Institute of Cancer Research, London, United Kingdom.
Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Matt Coffey
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Cancer Research UK Clinical Centre, St. James’ University Hospital, Leeds, United Kingdom.
Department of Ophthalmology and Ocular Oncology,
St George’s Hospital Medical School, Tooting, London, United Kingdom.
Oncolytics Biotech Inc., Calgary, Canada.
Department of Oncology, University of Surrey, Guildford, United Kingdom.
The Institute of Cancer Research, London, United Kingdom.
Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Hardev Pandha
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Cancer Research UK Clinical Centre, St. James’ University Hospital, Leeds, United Kingdom.
Department of Ophthalmology and Ocular Oncology,
St George’s Hospital Medical School, Tooting, London, United Kingdom.
Oncolytics Biotech Inc., Calgary, Canada.
Department of Oncology, University of Surrey, Guildford, United Kingdom.
The Institute of Cancer Research, London, United Kingdom.
Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - John Chester
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Cancer Research UK Clinical Centre, St. James’ University Hospital, Leeds, United Kingdom.
Department of Ophthalmology and Ocular Oncology,
St George’s Hospital Medical School, Tooting, London, United Kingdom.
Oncolytics Biotech Inc., Calgary, Canada.
Department of Oncology, University of Surrey, Guildford, United Kingdom.
The Institute of Cancer Research, London, United Kingdom.
Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Alan Melcher
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Cancer Research UK Clinical Centre, St. James’ University Hospital, Leeds, United Kingdom.
Department of Ophthalmology and Ocular Oncology,
St George’s Hospital Medical School, Tooting, London, United Kingdom.
Oncolytics Biotech Inc., Calgary, Canada.
Department of Oncology, University of Surrey, Guildford, United Kingdom.
The Institute of Cancer Research, London, United Kingdom.
Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kevin Harrington
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Cancer Research UK Clinical Centre, St. James’ University Hospital, Leeds, United Kingdom.
Department of Ophthalmology and Ocular Oncology,
St George’s Hospital Medical School, Tooting, London, United Kingdom.
Oncolytics Biotech Inc., Calgary, Canada.
Department of Oncology, University of Surrey, Guildford, United Kingdom.
The Institute of Cancer Research, London, United Kingdom.
Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Richard Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Cancer Research UK Clinical Centre, St. James’ University Hospital, Leeds, United Kingdom.
Department of Ophthalmology and Ocular Oncology,
St George’s Hospital Medical School, Tooting, London, United Kingdom.
Oncolytics Biotech Inc., Calgary, Canada.
Department of Oncology, University of Surrey, Guildford, United Kingdom.
The Institute of Cancer Research, London, United Kingdom.
Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
9
|
Effect of heat shock protein-90 (HSP90) and vascular endothelial growth factor (VEGF) on survival in acute lymphoblastic leukemia: an immunohistochemical study. Med Oncol 2010; 28:846-51. [PMID: 20422320 DOI: 10.1007/s12032-010-9533-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Accepted: 04/07/2010] [Indexed: 12/14/2022]
Abstract
BACKGROUND The significance of vascular endothelial growth factor (VEGF) and heat shock protein-90 (HSP90) has received only limited attention especially in acute lymphoblastic leukemia (ALL). In this study, we assessed expressions of HSP90 and VEGF in bone marrow samples of patients with ALL and effect of these expression quantities on the mean overall survival. PATIENTS AND METHODS Using immunohistochemical methods, we assessed expression of HSP90 and VEGF in 22 cases of ALL. RESULTS Expression of HSP90 was detected in 19/22 (86.4%) and 3/22 (13.6%) of patients with ALL, for strongly positive and moderate-weakly positive, respectively. Negative HSP90 expression was not detected in patients with ALL. Expression of HSP90 in patients with ALL and in control group were statistically significant (P<0.001), however, did not reflect the mean overall survival (P=0.910). Mean OS was evaluated 992±181 and 724.8±88.2 days for moderate-weak and high HSP90 expression, respectively. VEGF expressions were not significantly different between ALL and control groups (P<0.087). We did not find any relationship between HSP90 and VEGF expressions in bone marrow specimens of patients with ALL. CONCLUSION This study demonstrated that HSP90 expression grades in patients with ALL were significantly higher than that in controls and presence of strong HSP90 expression was associated with worse overall survival. VEGF expression in patients with ALL was not different from that in control samples. Determination HSP90 with immunohistochemical method in bone marrow can provide information about prognosis.
Collapse
|
10
|
Expression of VEGF and VEGF receptors in childhood precursor B-cell acute lymphoblastic leukemia evaluated by immunohistochemistry. J Pediatr Hematol Oncol 2009; 31:696-701. [PMID: 19707156 DOI: 10.1097/mph.0b013e3181b258df] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Perturbation in the expression and signaling pathways of vascular endothelial growth factor (VEGF) has been linked to pathogenesis of hematologic malignancies. We investigated the expression and clinical importance of VEGF and two of its receptors, VEGFR-1 and VEGFR-2, in childhood precursor B-cell acute lymphoblastic leukemia (pre-B ALL) by using immunohistochemistry. These angiogenic proteins were expressed in the majority of leukemic bone marrow samples. Notably, pre-B ALL patients had significantly increased expression of VEGFR-1 compared with no expression in the nonmalignant group, indicating a link between VEGFR-1 protein expression and pre-B ALL. These novel findings suggest that VEGFR-1 may have clinical importance in childhood pre-B ALL.
Collapse
|
11
|
Growth inhibition by tyrosine kinase inhibitors in mesothelioma cell lines. Eur J Cancer 2009; 45:1684-91. [PMID: 19318229 DOI: 10.1016/j.ejca.2009.02.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 02/17/2009] [Accepted: 02/23/2009] [Indexed: 11/24/2022]
Abstract
Clinical outcome following chemotherapy for malignant pleural mesothelioma is poor and improvements are needed. This preclinical study investigates the effect of five tyrosine kinase inhibitors (PTK787, ZD6474, ZD1839, SU6668 and SU11248) on the growth of three mesothelioma cell lines (NCI H226, NCI H28 and MSTO 211H), the presence of growth factor receptors and inhibition of their downstream signalling pathways. GI50 values were determined: ZD6474 and SU11248, mainly VEGFR2 inhibitors, gave the lowest GI50 across all cell lines (3.5-6.9 microM) whereas ZD1839 gave a GI50 in this range only in H28 cells. All cell lines were positive for EGFR, but only H226 cells were positive for VEGFR2 by Western blotting. ZD6474 and ZD1839 inhibited EGF-induced phosphorylation of EGFR, AKT and ERK, whereas VEGF-induced phosphorylation of VEGFR2 was completely inhibited with 0.1 microM SU11248. VEGFR2 was detected in tumour samples by immunohistochemistry. VEGFR2 tyrosine kinase inhibitors warrant further investigation in mesothelioma.
Collapse
|
12
|
Bando Y, Noguchi K, Kobayashi H, Yoshida N, Ishikawa I, Izumi Y. Cyclooxygenase-2-derived prostaglandin E2 is involved in vascular endothelial growth factor production in interleukin-1alpha-stimulated human periodontal ligament cells. J Periodontal Res 2009; 44:395-401. [PMID: 19210337 DOI: 10.1111/j.1600-0765.2008.01118.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND OBJECTIVE Prostaglandin E(2), which exerts its actions via EP receptors (EP1, EP2, EP3 and EP4), is a bioactive metabolite of arachidonic acid produced by cyclooxygenase-1 and/or cyclooxygenase-2. Interleukin-1alpha induces prostaglandin E(2) production via cyclooxygenase-2 in human periodontal ligament cells. Vascular endothelial growth factor is a key regulator of physiologic as well as pathologic angiogenesis and has been indicated to be involved in the pathology of periodontal diseases. In the present study, we investigated whether interleukin-1alpha induced vascular endothelial growth factor production in human periodontal ligament cells and whether cyclooxygenase-2-derived prostaglandin E(2) regulated interleukin-1alpha-induced vascular endothelial growth factor production. MATERIAL AND METHODS Human periodontal ligament cells were obtained from extracted teeth of periodontally healthy subjects. After pre-incubation with a nonselective cyclooxygenase-1/2 inhibitor, indomethacin or a selective cyclooxygenase-2 inhibitor (NS-398), periodontal ligament cells were treated with or without interleukin-1alpha, prostaglandin E(2), various EP receptor agonists and dibutyryl cAMP (a cAMP analogue). The levels of vascular endothelial growth factor and prostaglandin E(2) in the culture supernatant were measured by enzyme-linked immunosorbent assay. The vascular endothelial growth factor mRNA expression was evaluated by semiquantitative reverse transcription-polymerase chain reaction. RESULTS Interleukin-1alpha induced vascular endothelial growth factor production in a dose-dependent and time-dependent manner. The interleukin-1alpha-induced vascular endothelial growth factor mRNA and protein expression was inhibited to the same extent by indomethacin and NS-398. Indomethacin and NS-398 completely inhibited interleukin-1alpha-induced prostaglandin E(2) production. Exogenous prostaglandin E(2), butaprost (an EP2 receptor agonist) and dibutyryl cAMP abolished the inhibitory effect of indomethacin on interleukin-1alpha-induced vascular endothelial growth factor production. CONCLUSION We suggest that interleukin-1alpha induced vascular endothelial growth factor production via cyclooxygenase-2-derived prostaglandin E(2) in human periodontal ligament cells. The interleukin-1alpha/prostaglandin E(2) pathway might regulate vascular endothelial growth factor production in periodontal lesions.
Collapse
Affiliation(s)
- Y Bando
- Periodontology, Department of Hard Tissue Engineering, Tokyo Medical and Dental University, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
13
|
Poyer F, Coquerel B, Pegahi R, Cazin L, Norris V, Vannier JP, Lamacz M. Secretion of MMP-2 and MMP-9 induced by VEGF autocrine loop correlates with clinical features in childhood acute lymphoblastic leukemia. Leuk Res 2008; 33:407-17. [PMID: 18829111 DOI: 10.1016/j.leukres.2008.08.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Revised: 07/30/2008] [Accepted: 08/23/2008] [Indexed: 10/21/2022]
Abstract
In children with acute lymphoblastic leukemia (ALL), leukemic cells express several members of the VEGF family and the three VEGF receptors which, via an autocrine loop are responsible for secretion of MMP-2/-9. MMP activity and the presence of elements of the autocrine loop are correlated with clinical and prognostic parameters as follows: i) high basal MMP-9 activity with tumoral syndrome, ii) MMP-2 activity with treatment failure, iii) VEGFR-1/-3 co-expression with high hemoglobin level and iv) expression of the VEGF-A 121 isoform and favorable response to treatment. These data implicate autocrine VEGF-induced secretion of MMP-2/-9 in the physiopathology of childhood ALL.
Collapse
Affiliation(s)
- Florent Poyer
- Groupe de Recherche sur le Micro-Environnement et le Renouvellement Cellulaire Intégré M.E.R.C.I., Faculté de Médecine-Pharmacie, Université de Rouen; 22 bd Gambetta, 76183 Rouen, France
| | | | | | | | | | | | | |
Collapse
|
14
|
Styczynski J, Czyzewski K, Wysocki M. Ex vivo activity of thalidomide in childhood acute leukemia. Leuk Lymphoma 2007; 47:1123-8. [PMID: 16840205 DOI: 10.1080/10428190500467891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Thalidomide is a drug with anti-angiogenic, anti-inflammatory, immunomodulatory and anti-cancer properties that were found to inhibit the production of TNF-alpha in vitro, stimulate reactive oxygen species production, and inhibit VEGFR in acute leukemias. Ex vivo activity of thalidomide as a single agent and in combination with prednisolone or cytarabine in childhood acute leukemias was analyzed. Forty samples of childhood acute lymphoblastic leukemia (ALL) and 13 acute myeloid leukemia (AML) were tested for cytotoxicity by the MTT assay and cell cycle phases by flow cytometry. Control studies were performed on 9 samples of normal lymphocytes and 4 cell lines. A weak anti-leukemic activity of thalidomide against childhood leukemic samples was observed. However, in the presence of thalidomide, cytotoxicity of prednisolone or cytarabine, increased 3.3-fold and 2.7-fold, respectively, in childhood ALL but was not changed in AML. Thalidomide increased apoptosis in lymphoblasts, and modulated cell cycle arrest caused by prednisolone but not cytarabine in childhood acute lymphoblastic leukemia samples. Thalidomide potentiated ex vivo sensitivity of childhood ALL cells to prednisolone and cytarabine, while no sensitization effect was observed in AML cells.
Collapse
Affiliation(s)
- Jan Styczynski
- Department of Pediatric Hematology and Oncology, Ludwik Rydygier's Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland.
| | | | | |
Collapse
|
15
|
Adair A, Mitchell DR, Kipari T, Qi F, Bellamy COC, Robertson F, Hughes J, Marson LP. Peritubular capillary rarefaction and lymphangiogenesis in chronic allograft failure. Transplantation 2007; 83:1542-50. [PMID: 17589335 DOI: 10.1097/01.tp.0000266689.93615.cd] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Chronic renal allograft failure is a common and multifactorial but incompletely understood process with no effective treatment strategy. METHODS We used immunohistochemistry to evaluate changes in density and turnover (proliferation) of the microvasculature and lymphatic vessels in endstage human transplant nephrectomies and control tissue derived from macroscopically normal areas of native nephrectomy specimens removed for renal carcinoma. We also examined the expression of angiogenic and lymphangiogenic growth factors in the associated inflammatory infiltrate. RESULTS Endstage allografts showed reduced microvascular density in cortex and medulla compared with controls (P<0.0001), despite the presence of endothelial cell proliferation. However, the grafts also showed new lymphatic vessels in the tubulointerstitium, not evident in controls, and which appeared to be functional with luminal macrophages. Double labeling studies showed a subpopulation of the graft-infiltrating macrophages to be immunopositive for inducible nitric oxide synthase or vascular endothelial growth factor-C (a lymphatic-specific growth factor). B cells also strongly expressed the inflammatory and angiogenic cytokine vascular endothelial growth factor A. CONCLUSIONS The present results identify contrasting changes in the microanatomy of vascular and lymphatic beds in endstage renal allografts associated with subpopulations of infiltrating macrophages and B cells that potentially regulate some of these changes. These cells and processes could become a new therapeutic target in chronic allograft failure.
Collapse
Affiliation(s)
- Anya Adair
- Phagocyte Laboratory, MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
16
|
El-Obeid A, Hassib A, Pontén F, Westermark B. Effect of herbal melanin on IL-8: a possible role of Toll-like receptor 4 (TLR4). Biochem Biophys Res Commun 2006; 344:1200-6. [PMID: 16650380 DOI: 10.1016/j.bbrc.2006.04.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2006] [Accepted: 04/05/2006] [Indexed: 11/25/2022]
Abstract
The production of IL-8 can be induced by LPS via TLR4 signaling pathway. In this study, we tested the effect of a herbal melanin (HM) extract, from black cumin seeds (Nigella sativa L.), on IL-8 production. We used HM and LPS in parallel to induce IL-8 production by THP-I, PBMCs, and TLR4-transfected HEK293 cells. Both HM and LPS induced IL-8 mRNA expression and protein production in THP-1 and PBMCs. On applying similar treatment to HEK293 cells that express TLR4, MD2, and CD14, both HM and LPS significantly induced IL-8 protein production. We have also demonstrated that HM and LPS had identical effects in terms of IL-8 stimulation by HEK293 transfected with either TLR4 or MD2-CD14. Melanin extracted from N. sativa L. mimics the action of LPS in the induction of IL-8 by PBMC and the other used cell lines. Our results suggest that HM may share a signaling pathway with LPS that involves TLR4.
Collapse
Affiliation(s)
- Adila El-Obeid
- Department of Genetics and Pathology, Rudbeck Laboratory, Uppsala University Hospital, SE-751 85 Uppsala, Sweden.
| | | | | | | |
Collapse
|
17
|
El-Obeid A, Al-Harbi S, Al-Jomah N, Hassib A. Herbal melanin modulates tumor necrosis factor alpha (TNF-alpha), interleukin 6 (IL-6) and vascular endothelial growth factor (VEGF) production. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2006; 13:324-33. [PMID: 16635740 DOI: 10.1016/j.phymed.2005.03.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2004] [Accepted: 03/17/2005] [Indexed: 05/08/2023]
Abstract
Recent studies have indicated that cytokines can enhance immunogenicity and promote tumor regression. However, the means for modulating cytokine production are not yet fully investigated. In this study we report the effects of a herbal melanin, extracted from Nigella sativa L., on the production of three cytokines [tumor necrosis factor alpha (TNF-alpha), interleukin 6 (IL-6) and vascular endothelial growth factor (VEGF)], by human monocytes, total peripheral blood mononuclear cells (PBMC) and THP-1 cell line. Cells were treated with variable concentrations of melanin and the expression of TNF-alpha, IL-6 and VEGF mRNA in cell lysates and secretion of proteins in the supernatants were detected by RT-PCR and ELISA. Melanin induced TNF-alpha, IL-6 and VEGF mRNA expression by the monocytes, PBMC and THP-1 cell line. On the protein level, melanin significantly induced TNF-alpha and IL-6 protein production and inhibited VEGF production by monocytes and PBMC. In the THP-1 cell line melanin induced production of all three cytokine proteins. These observations raise the prospects of using N. sativa L. melanin for treatment of diseases associated with imbalanced cytokine production and for enhancing cancer and other immunotherapies.
Collapse
Affiliation(s)
- A El-Obeid
- Biological Repository Center, Department of Molecular Pathology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | | | | | | |
Collapse
|
18
|
Dong Y, Qian J, Ibrahim R, Berzofsky JA, Khleif SN. Identification of H-2Db-Specific CD8+ T-Cell Epitopes From Mouse VEGFR2 That Can Inhibit Angiogenesis and Tumor Growth. J Immunother 2006; 29:32-40. [PMID: 16365598 DOI: 10.1097/01.cji.0000175494.13476.56] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2/KDR) plays a crucial role in tumor-associated angiogenesis and vascularization. It has been established that monoclonal antibodies against VEGFR2 can inhibit angiogenesis. In this study, two naturally processed CD8 T-cell epitopes (VILTNPISM and FSNSTNDILI) were identified from murine KDR. Cytotoxic T lymphocytes targeting endothelial cells could be directly monitored by KDR2 and KDR3 Elispots or major histocompatibility complex class I tetramer staining. Immunization with these two peptides effectively reduced angiogenesis and inhibited tumor growth in mouse models. Thus, vaccination with KDR peptides alone or in combination with other anti-angiogenesis agents may afford a novel immunotherapy for inhibition of tumor growth.
Collapse
Affiliation(s)
- Yujun Dong
- The Vaccine Branch, The Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20889, USA
| | | | | | | | | |
Collapse
|
19
|
Coppola S, Narciso L, Feccia T, Bonci D, Calabrò L, Morsilli O, Gabbianelli M, De Maria R, Testa U, Peschle C. Enforced expression of KDR receptor promotes proliferation, survival and megakaryocytic differentiation of TF1 progenitor cell line. Cell Death Differ 2005; 13:61-74. [PMID: 15962006 DOI: 10.1038/sj.cdd.4401698] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) receptor-2/kinase insert domain-containing receptor (KDR) is expressed in primitive hematopoietic cells, in megakaryocytes and platelets. In primitive hematopoiesis KDR mediates cell survival via autocrine VEGF, while its effect on cell growth and differentiation has not been elucidated. We induced enforced KDR expression in the granulocyte macrophage-colony-stimulating factor (GM-CSF)-dependent TF1 progenitor cell line (TF1-KDR), treated the cells with VEGF and analyzed their response. In GM-CSF-deprived cells, VEGF induces cell proliferation and protection against apoptosis, followed by enhanced expression of megakaryocytic (MK) markers. Combined with GM-CSF, VEGF induces a mild proliferative stimulus, followed by cell adherence, accumulation in G0/G1, massive MK differentiation and Fas-mediated apoptosis. Accordingly, we observed that MK-differentiating cells, derived from hematopoietic progenitors, produce VEGF, express KDR, inhibition of which reduces MK differentiation, indicating a key role of KDR in megakaryopoiesis. In conclusion, TF1-KDR cells provide a reliable model to investigate the biochemical and molecular mechanisms underlying hematopoietic progenitor proliferation, survival and MK differentiation.
Collapse
Affiliation(s)
- S Coppola
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Hansen T, Gaumann A, Ghalibafian M, Höferlin A, Heintz A, Kirkpatrick CJ. Haemangiopericytoma of the thyroid gland in combination with Hashimoto?s disease. Virchows Arch 2004; 445:315-9. [PMID: 15243738 DOI: 10.1007/s00428-004-1066-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2004] [Accepted: 05/29/2004] [Indexed: 01/06/2023]
Abstract
We present a hitherto unique case of haemangiopericytoma (HP) of the thyroid gland in a 15-year-old female patient suffering from Hashimoto's disease for several months. Since angiogenesis has been discussed to play a major role in both diseases, we examined the expression of vascular endothelial growth factor (VEGF), VEGF receptors (VEGFRs) and platelet-derived growth factor receptors (PDGFRs). Most interestingly, strong expression of PDGFR alpha and beta was found in spindle-shaped tumour cells and tumour vessels in HP, while VEGF and VEGFR type I and -II were negative in these regions. In contrast, VEGF was expressed in the lymphoid infiltrate of Hashimoto's disease. Since PDGFR-beta is commonly expressed in pericytes, we suggest that the strong expression discovered in this study further supports the view that HP is derived from pericytes. The combination of HP and Hashimoto's disease is most probably a coincidental event. However, this case confirms previous reports demonstrating that in patients with Hashimoto's disease different neoplasias can occur.
Collapse
Affiliation(s)
- T Hansen
- Institute of Pathology, Johannes Gutenberg, University of Mainz, Langenbeckstrasse 1, 55101 Mainz, Germany.
| | | | | | | | | | | |
Collapse
|