1
|
Rajkumari S, Singh J, Agrawal U, Agrawal S. Myeloid-derived suppressor cells in cancer: Current knowledge and future perspectives. Int Immunopharmacol 2024; 142:112949. [PMID: 39236460 DOI: 10.1016/j.intimp.2024.112949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024]
Abstract
MDSCs (myeloid-derived suppressor cells) are crucial for immune system evasion in cancer. They accumulate in peripheral blood and tumor microenvironment, suppressing immune cells like T-cells, natural killer cells and dendritic cells. They promote tumor angiogenesis and metastasis by secreting cytokines and growth factors and contribute to a tumor-promoting environment. The accumulation of MDSCs in cancer patients has been linked to poor prognosis and resistance to various cancer therapies. Targeting MDSCs and their immunosuppressive mechanisms may improve treatment outcomes and enhance immune surveillance by developing drugs that inhibit MDSC function, by preventing their accumulation and by disrupting the tumor-promoting environment. This review presents a detailed overview of the MDSC research in cancer with regulation of their development and function. The relevance of MDSC as a prognostic and predictive biomarker in different types of cancers, along with recent advancements on the therapeutic approaches to target MDSCs are discussed in detail.
Collapse
Affiliation(s)
- Sunanda Rajkumari
- ICMR National Institute of Medical Statistics, Ansari Nagar, New Delhi 110029, India
| | - Jaspreet Singh
- ICMR National Institute of Pathology, Safdarjung Hospital Campus, Ansari Nagar, New Delhi 110029, India
| | - Usha Agrawal
- Asian Institute of Public Health University (AIPH) University, 1001 Haridamada, Jatani, Near IIT Bhubaneswar, Bhubaneswar 751002, India
| | - Sandeep Agrawal
- Discovery Research Division, Indian Council of Medical Research, Ansari Nagar, New Delhi 110029, India.
| |
Collapse
|
2
|
Sadighi S, Sahinoglu E, Kubba AH, Patel J, Hosseini F, Shafiee MA, Qureshi B. Impact of Serum Ferritin and Iron Overload on Acute Myeloid Leukemia Outcomes: A Systematic Review and Meta-Analysis. Asian Pac J Cancer Prev 2024; 25:2951-2962. [PMID: 39205594 PMCID: PMC11495433 DOI: 10.31557/apjcp.2024.25.8.2951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE To evaluate the iron overload among individuals with acute myeloid leukemia (AML) who have not received red blood cell transfusions. METHODS A comprehensive search was conducted in Embase, PubMed, PubMed Central, Web of Science, NIH, and Blood Library databases up to September 2023. The search strategy included keywords related to AML, iron overload, serum ferritin, survival, outcomes, and inflammation. Manual searches through included articles and relevant references were also performed. From 1650 initial articles, 16 studies involving 8752 patients met the inclusion criteria for systematic review. Statistical analysis used hazard ratios (HR) and confidence intervals (CI). Results: The systematic review and meta-analysis revealed a statistically significant association between high serum ferritin (SF) levels and poor outcomes in AML patients before starting chemotherapy. Elevated SF levels (>1000 mg/L) were associated with lower overall survival (OS) and event-free survival (EFS) (HR for OS: 1.99, 95% CI: 1.48-2.66; HR for EFS: 2.29, 95% CI: 1.73-3.05). Elevated SF levels were inversely correlated with the gradual onset of infections, indicating an increased risk of early mortality (p<0.05). CONCLUSION Elevated serum ferritin levels are significantly associated with poor outcomes in AML patients before treatment initiation. These findings highlight the importance of monitoring iron levels in these patients to improve prognostic assessments and treatment strategies.
Collapse
Affiliation(s)
- Sanambar Sadighi
- Hematology-Oncology Department, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | | | | | | | |
Collapse
|
3
|
Potre C, Borsi E, Potre O, Ionita I, Samfireag M, Costachescu D, Secosan C, Lazar S, Ristescu AI. A Systematic Review Assessing the Impact of Vitamin D Levels on Adult Patients with Lymphoid Malignancies. Curr Oncol 2023; 30:4351-4364. [PMID: 37185444 PMCID: PMC10137018 DOI: 10.3390/curroncol30040331] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Vitamin D deficiency has been correlated with various conditions, including the risk of developing lymphoid malignancies. This systematic review aimed to assess the association between vitamin D levels at diagnosis of lymphoid malignancies, patient outcomes, and survival. A systematic review was conducted, encompassing 15 studies published until January 2023, involving 4503 patients, examining the relationship between vitamin D and lymphoid cancers. The median age of the patients was 56.5 years, with a median follow-up duration of approximately 36 months across studies. The overall median vitamin D level at initial measurement was 20.4 ng/mL, while a <20 ng/mL threshold was used to define vitamin D insufficiency. The results demonstrated significant associations between vitamin D levels and patient outcomes in several lymphoid malignancies, with a pooled risk in disease progression of 1.93 and a pooled hazard ratio of 2.06 for overall survival in patients with 25-(OH)D levels below the normal threshold of 20 ng/mL. Among findings, it was demonstrated that supplemental vitamin D improves the chemosensitivity of tumors by reducing the rate of tumor growth compared with vitamin D or chemotherapy alone. Vitamin D had a protective effect for patients with DLBCL under R-CHOP treatment, while vitamin D insufficiency was associated with the impairment of rituximab treatment and showed worse clinical outcomes in chimeric antigen receptor T-cell (CAR-T) recipients. Although one study found no association between vitamin D deficiency and the cause of death, most associated vitamin D insufficiency with early clinical failure and lower survival probability. In conclusion, his systematic review highlights the importance of vitamin D levels in the prognosis and survival of patients with lymphoid malignancies. Further research is needed to better understand the underlying mechanisms and explore the potential benefits of vitamin D supplementation in managing these cancers.
Collapse
Affiliation(s)
- Cristina Potre
- Department of Internal Medicine, Discipline of Hematology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Ema Borsi
- Department of Internal Medicine, Discipline of Hematology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Ovidiu Potre
- Department of Internal Medicine, Discipline of Hematology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Ioana Ionita
- Department of Internal Medicine, Discipline of Hematology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Miruna Samfireag
- Department of Internal Medicine, Discipline of Clinical Practical Skills, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Dan Costachescu
- Department of Orthopedics-Traumatology, Urology, Radiology, and Medical Imaging, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Cristina Secosan
- Department of Obstetrics and Gynecology, Discipline of Obstetrics and Gynecology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Sandra Lazar
- Second Department of Internal Medicine, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Anca Irina Ristescu
- Discipline of Anesthesia and Intensive Care, School of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, University Street 16, 700115 Iasi, Romania
- Department of Anesthesia and Intensive Care, Regional Institute of Oncology, General Henri Mathias Berthelot 2-4, 700483 Iasi, Romania
| |
Collapse
|
4
|
Zhao Y, Du J, Shen X. Targeting myeloid-derived suppressor cells in tumor immunotherapy: Current, future and beyond. Front Immunol 2023; 14:1157537. [PMID: 37006306 PMCID: PMC10063857 DOI: 10.3389/fimmu.2023.1157537] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/06/2023] [Indexed: 03/19/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are one of the major negative regulators in tumor microenvironment (TME) due to their potent immunosuppressive capacity. MDSCs are the products of myeloid progenitor abnormal differentiation in bone marrow, which inhibits the immune response mediated by T cells, natural killer cells and dendritic cells; promotes the generation of regulatory T cells and tumor-associated macrophages; drives the immune escape; and finally leads to tumor progression and metastasis. In this review, we highlight key features of MDSCs biology in TME that are being explored as potential targets for tumor immunotherapy. We discuss the therapies and approaches that aim to reprogram TME from immunosuppressive to immunostimulatory circumstance, which prevents MDSC immunosuppression activity; promotes MDSC differentiation; and impacts MDSC recruitment and abundance in tumor site. We also summarize current advances in the identification of rational combinatorial strategies to improve clinical efficacy and outcomes of cancer patients, via deeply understanding and pursuing the mechanisms and characterization of MDSCs generation and suppression in TME.
Collapse
Affiliation(s)
- Yang Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Junfeng Du
- Department of General Surgery, The 7th Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- *Correspondence: Junfeng Du, ; Xiaofei Shen,
| | - Xiaofei Shen
- Department of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Junfeng Du, ; Xiaofei Shen,
| |
Collapse
|
5
|
Marchwicka A, Nowak U, Grembowska A, Jakuszak A, Poręba P, Marcinkowska E. Overexpressed fibroblast growth factor receptors increase 1,25-dihydroxyvitamin D-dependent differentiation of acute myeloid leukemia cells. J Steroid Biochem Mol Biol 2022; 224:106173. [PMID: 36031072 DOI: 10.1016/j.jsbmb.2022.106173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/09/2022] [Accepted: 08/23/2022] [Indexed: 11/23/2022]
Abstract
Many malignancies are driven by mutations within the gene for fibroblast growth factor receptor 1 (FGFR1). Previously, we have shown that signal transduction from the FOP2-FGFR1 fusion protein in acute myeloid leukemia KG1 cells is responsible for a low level of expression of the vitamin D receptor gene. In this paper, we address whether other fibroblast growth factor receptors regulate the vitamin D receptor (VDR) gene. We used the human myeloid leukemia U937 and HL60 cells, the bone cancer cell line U2OS, and cell transfection methods to answer the question. For myeloid leukemia cells, overexpression of FGFRs 1-3 genes caused a shift towards monocytic differentiation; this was extracellular regulated kinase (Erk) 1,2-dependent. Overexpression of FGFRs 1-3 genes also upregulated expression of the VDR gene, further sensitizing these cells to 1,25-dihydroxyvitamin D-induced monocyte differentiation. When we increased expression in bone cells, fibroblast growth factor receptors did not upregulate VDR gene expression, nor influence the activity of VDR. Fibroblast growth factor receptors are overexpressed in many neoplasms. Therefore, it may be reasonable to use vitamin D analogs to treat these cancers, to activate VDR and drive cell differentiation.
Collapse
Affiliation(s)
- Aleksandra Marchwicka
- Department of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Urszula Nowak
- Department of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Anna Grembowska
- Department of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Agnieszka Jakuszak
- Department of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Piotr Poręba
- Department of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Ewa Marcinkowska
- Department of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| |
Collapse
|
6
|
Morales-Guadarrama G, Méndez-Pérez EA, García-Quiroz J, Avila E, Larrea F, Díaz L. AZD4547 and calcitriol synergistically inhibited BT-474 cell proliferation while modified stemness and tumorsphere formation. J Steroid Biochem Mol Biol 2022; 223:106132. [PMID: 35659529 DOI: 10.1016/j.jsbmb.2022.106132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/02/2022] [Accepted: 05/22/2022] [Indexed: 10/18/2022]
Abstract
Fibroblast growth factor receptor (FGFR) overamplification/activation in cancer leads to increased cell proliferation. AZD4547, a FGFR selective inhibitor, hinders breast cancer cells growth. Although luminal B breast tumors may respond to chemotherapy and endocrine therapy, this subtype is associated with poor prognosis, inadequate response and/or acquired drug resistance. Calcitriol, the vitamin D most active metabolite, exerts anti-neoplastic effects and enhances chemotherapeutic drugs activity. In this study, we sought to decrease the concentration of AZD4547 needed to inhibit the luminal-B breast cancer cell line BT-474 proliferation by its combination with calcitriol. Anti-proliferative inhibitory concentrations, combination index and dose-reduction index were analyzed from Sulforhodamine B assays. Western blot and qPCR were used to study FGFR molecular targets. The compound's ability to inhibit BT-474 cells tumorigenic capacity was assessed by tumorspheres formation. Results: BT-474 cells were dose-dependently growth-inhibited by calcitriol and AZD4547 (IC50 = 2.9 nM and 3.08 μM, respectively). Calcitriol at 1 nM synergistically improved AZD4547 antiproliferative effects, allowing a 2-fold AZD4547 dose-reduction. Mechanistically, AZD4547 downregulated p-FGFR1, p-Akt and tumorsphere formation. Calcitriol also decreased tumorspheres, while induced cell differentiation. Both compounds inhibited MYC and CCND1 expression, as well as ALDH, a stemness marker that positively correlated with FGFR1 and negatively with VDR expression in breast cancer transcriptomic data. In conclusion, the drugs impaired self-aggregation capacity, reduced stemness features, induced cell-differentiation and when combined, synergistically inhibited cell proliferation. Overall, our results suggest that calcitriol, at low pharmacological doses, may be a suitable candidate to synergize AZD4547 effects in luminal B breast tumors, allowing to reduce dose and adverse effects.
Collapse
Affiliation(s)
- Gabriela Morales-Guadarrama
- Departamento de Biología de la Reproducción, Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan 14080, Ciudad de México, Mexico; Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| | - Edgar A Méndez-Pérez
- Departamento de Biología de la Reproducción, Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan 14080, Ciudad de México, Mexico.
| | - Janice García-Quiroz
- Departamento de Biología de la Reproducción, Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan 14080, Ciudad de México, Mexico.
| | - Euclides Avila
- Departamento de Biología de la Reproducción, Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan 14080, Ciudad de México, Mexico.
| | - Fernando Larrea
- Departamento de Biología de la Reproducción, Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan 14080, Ciudad de México, Mexico.
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción, Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan 14080, Ciudad de México, Mexico.
| |
Collapse
|
7
|
Hino C, Pham B, Park D, Yang C, Nguyen MH, Kaur S, Reeves ME, Xu Y, Nishino K, Pu L, Kwon SM, Zhong JF, Zhang KK, Xie L, Chong EG, Chen CS, Nguyen V, Castillo DR, Cao H. Targeting the Tumor Microenvironment in Acute Myeloid Leukemia: The Future of Immunotherapy and Natural Products. Biomedicines 2022; 10:biomedicines10061410. [PMID: 35740430 PMCID: PMC9219790 DOI: 10.3390/biomedicines10061410] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) plays an essential role in the development, proliferation, and survival of leukemic blasts in acute myeloid leukemia (AML). Within the bone marrow and peripheral blood, various phenotypically and functionally altered cells in the TME provide critical signals to suppress the anti-tumor immune response, allowing tumor cells to evade elimination. Thus, unraveling the complex interplay between AML and its microenvironment may have important clinical implications and are essential to directing the development of novel targeted therapies. This review summarizes recent advancements in our understanding of the AML TME and its ramifications on current immunotherapeutic strategies. We further review the role of natural products in modulating the TME to enhance response to immunotherapy.
Collapse
Affiliation(s)
- Christopher Hino
- Department of Internal Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.H.); (B.P.); (K.N.); (L.P.); (S.M.K.)
| | - Bryan Pham
- Department of Internal Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.H.); (B.P.); (K.N.); (L.P.); (S.M.K.)
| | - Daniel Park
- Department of Internal Medicine, School of Medicine, University of California San Francisco–Fresno, Fresno, CA 93701, USA;
| | - Chieh Yang
- Department of Internal Medicine, School of Medicine, University of California Riverside, Riverside, CA 92521, USA;
| | - Michael H.K. Nguyen
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
| | - Simmer Kaur
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
| | - Mark E. Reeves
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
| | - Yi Xu
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
| | - Kevin Nishino
- Department of Internal Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.H.); (B.P.); (K.N.); (L.P.); (S.M.K.)
| | - Lu Pu
- Department of Internal Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.H.); (B.P.); (K.N.); (L.P.); (S.M.K.)
| | - Sue Min Kwon
- Department of Internal Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.H.); (B.P.); (K.N.); (L.P.); (S.M.K.)
| | - Jiang F. Zhong
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, USA;
| | - Ke K. Zhang
- Department of Nutrition, Texas A&M University, College Station, TX 77030, USA; (K.K.Z.); (L.X.)
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Linglin Xie
- Department of Nutrition, Texas A&M University, College Station, TX 77030, USA; (K.K.Z.); (L.X.)
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Esther G. Chong
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
| | - Chien-Shing Chen
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
| | - Vinh Nguyen
- Department of Biology, University of California Riverside, Riverside, CA 92521, USA;
| | - Dan Ran Castillo
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
- Correspondence: (D.R.C.); (H.C.)
| | - Huynh Cao
- Department of Oncology/Hematology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (M.H.K.N.); (S.K.); (M.E.R.); (Y.X.); (E.G.C.); (C.-S.C.)
- Correspondence: (D.R.C.); (H.C.)
| |
Collapse
|
8
|
Vieco-Martí I, López-Carrasco A, de la Cruz-Merino L, Noguera R, Álvaro Naranjo T. The complexity of cancer immunotherapy illustrated through skin tumors. Int J Biol Markers 2022; 37:113-122. [PMID: 35473449 DOI: 10.1177/03936155221088884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Skin tumours are among the cancer types most sensitive to immunotherapy, due to their unique immunogenic features including skin-associated lymphoid tissue, high mutational load, overexpression of tumour antigens, and high frequency of viral antigens. Despite this high immunotherapy response rate, however, ultimately most skin tumours develop similar treatment resistance to most other malignant tumours, which highlights the need for in-depth study of mechanisms of response and resistance to immunotherapy. METHODS A bibliographic review of the most recent publications regarding currently in use and emerging biomarkers on skin tumors has been done. RESULTS Predictive biomarkers of treatment response, biomarkers that warn of possible resistance, and emerging markers, the majority of a systemic nature, are described. Including factors affecting not only genomics, but also the immune system, nervous system, microbiota, tumour microenvironment, metabolism and stress. CONCLUSIONS For accurate diagnosis of tumour type, knowledge of its functional mechanisms and selection of a comprehensive therapeutic protocol, this inclusive view of biology, health and disease is fundamental. This field of study could also become a valuable source of practical information applicable to other areas of oncology and immunotherapy.
Collapse
Affiliation(s)
- I Vieco-Martí
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain.,Centro de investigación biomédica en red de cáncer (CIBERONC), Madrid, Spain
| | - A López-Carrasco
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain.,Centro de investigación biomédica en red de cáncer (CIBERONC), Madrid, Spain
| | - L de la Cruz-Merino
- Departament of Oncology, Hospital Universitario Virgen Macarena, Seville, Spain
| | - R Noguera
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain.,Centro de investigación biomédica en red de cáncer (CIBERONC), Madrid, Spain
| | - T Álvaro Naranjo
- Centro de investigación biomédica en red de cáncer (CIBERONC), Madrid, Spain.,Department of Pathology, Hospital de Tortosa Verge de la Cinta, Catalan Institute of Health, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tortosa, Spain.,Department of Morphological Science, Medical School, Rovira i Virgili University, Reus, Spain
| |
Collapse
|
9
|
Hultmark S, Baudet A, Schmiderer L, Prabhala P, Palma-Tortosa S, Sandén C, Fioretos T, Sasidharan R, Larsson C, Lehmann S, Juliusson G, Ek F, Magnusson M. Combinatorial molecule screening identified a novel diterpene and the BET inhibitor CPI-203 as differentiation inducers of primary acute myeloid leukemia cells. Haematologica 2021; 106:2566-2577. [PMID: 32855276 PMCID: PMC8485661 DOI: 10.3324/haematol.2020.249177] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Indexed: 12/24/2022] Open
Abstract
Combination treatment has proven effective for patients with acute promyelocytic leukemia, exemplifying the importance of therapy targeting multiple components of oncogenic regulation for a successful outcome. However, recent studies have shown that the mutational complexity of acute myeloid leukemia (AML) precludes the translation of molecular targeting into clinical success. Here, as a complement to genetic profiling, we used unbiased, combinatorial in vitro drug screening to identify pathways that drive AML and to develop personalized combinatorial treatments. First, we screened 513 natural compounds on primary AML cells and identified a novel diterpene (H4) that preferentially induced differentiation of FLT3 wild-type AML, while FLT3-ITD/mutations conferred resistance. The samples responding to H4, displayed increased expression of myeloid markers, a clear decrease in the nuclear-cytoplasmic ratio and the potential of re-activation of the monocytic transcriptional program reducing leukemia propagation in vivo. By combinatorial screening using H4 and molecules with defined targets, we demonstrated that H4 induces differentiation by the activation of the protein kinase C (PKC) signaling pathway, and in line with this, activates PKC phosphorylation and translocation of PKC to the cell membrane. Furthermore, the combinatorial screening identified a bromo- and extra-terminal domain (BET) inhibitor that could further improve H4-dependent leukemic differentiation in FLT3 wild-type monocytic AML. These findings illustrate the value of an unbiased, multiplex screening platform for developing combinatorial therapeutic approaches for AML.
Collapse
Affiliation(s)
- Simon Hultmark
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Sweden
| | - Aurélie Baudet
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Sweden
| | - Ludwig Schmiderer
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Sweden
| | - Pavan Prabhala
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Sara Palma-Tortosa
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Carl Sandén
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Thoas Fioretos
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | | | - Christer Larsson
- Division of Translational Cancer Research, Lund University, Lund, Sweden
| | - Sören Lehmann
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Gunnar Juliusson
- Department of Hematology, Skane University Hospital, Lund, Sweden
| | - Fredrik Ek
- Chemical Biology and Therapeutics, Lund University, Lund, Sweden
| | - Mattias Magnusson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Sweden
| |
Collapse
|
10
|
Kazim N, Yen A. Role for Fgr and Numb in retinoic acid-induced differentiation and G0 arrest of non-APL AML cells. Oncotarget 2021; 12:1147-1164. [PMID: 34136084 PMCID: PMC8202776 DOI: 10.18632/oncotarget.27969] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/03/2021] [Indexed: 12/15/2022] Open
Abstract
Retinoic acid (RA) is a fundamental regulator of cell cycle and cell differentiation. Using a leukemic patient-derived in vitro model of a non-APL AML, we previously found that RA evokes activation of a macromolecular signaling complex, a signalosome, built of numerous MAPK-pathway-related signaling molecules; and this signaling enabled Retinoic-Acid-Response-Elements (RAREs) to regulate gene expression that results in cell differentiation/cell cycle arrest. Toward mechanistic insight into the nature of this novel signaling, we now find that the NUMB cell fate determinant protein is an apparent scaffold for the signalosome. Numb exists in the cell bound to an ensemble of signalosome molecules, including Raf, Lyn, Slp-76, and Vav. Addition of RA induces the expression of Fgr. Fgr binds NUMB, which is associated with (p-tyr)phosphorylation of NUMB and enhanced NUMB-binding and (p-tyr)phosphorylation of select signalosome components, thereby betraying signalosome activation. Signalosome activation is associated with cell differentiation along the myeloid lineage and G1/0 cell cycle arrest. If RA-induced Fgr expression is ablated by a CRISPR-KO; then the RA-induced (p-tyr) phosphorylation of NUMB and enhanced NUMB-binding and (p-tyr)phosphorylation of select signalosome components are lost. The cells now fail to undergo RA-induced differentiation or G1/0 arrest. In sum we find that NUMB acts as a scaffold for a signaling machine that functions to propel RA-induced differentiation and G1/0 arrest, and that Fgr binding to NUMB turns the function on. The Numb fate determinant protein thus appears to regulate the retinoic acid embryonic morphogen using the Fgr Src-Family-Kinase. These mechanistic insights suggest therapeutic targets for a hitherto incurable AML.
Collapse
Affiliation(s)
- Noor Kazim
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Andrew Yen
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
11
|
Tóth E, Erdődi F, Kiss A. Myosin Phosphatase Is Implicated in the Control of THP-1 Monocyte to Macrophage Differentiation. Int J Mol Sci 2021; 22:ijms22052516. [PMID: 33802280 PMCID: PMC7959147 DOI: 10.3390/ijms22052516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 01/22/2023] Open
Abstract
Monocyte to macrophage differentiation is characterized by the activation of various signal transduction pathways, which may be modulated by protein phosphorylation; however, the impact of protein kinases and phosphatases is not well understood yet. It has been demonstrated that actomyosin rearrangement during macrophage differentiation is dependent on Rho-associated protein kinase (ROCK). Myosin phosphatase (MP) target subunit-1 (MYPT1) is one of the major cellular substrates of ROCK, and MP is often a counter enzyme of ROCK; therefore, MP may also control macrophage differentiation. Changes in MP activity and the effects of MP activation were studied on PMA or l,25(OH)2D3-induced differentiation of monocytic THP-1 cells. During macrophage differentiation, phosphorylation of MYPT1 at Thr696 and Thr853 increased significantly, resulting in inhibition of MP. The ROCK inhibitor H1152 and the MP activator epigallocatechin-3-gallate (EGCG) attenuated MYPT1 phosphorylation and concomitantly decreased the extent of phosphorylation of 20 kDa myosin light chain. H1152 and EGCG pretreatment also suppressed the expression of CD11b and weakened the PMA-induced adherence of the cells. Our results indicate that MP activation/inhibition contributes to the efficacy of monocyte to macrophage differentiation, and this enzyme may be a target for pharmacological interventions in the control of disease states that are affected by excessive macrophage differentiation.
Collapse
Affiliation(s)
- Emese Tóth
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
- MTA-DE Cell Biology and Signalling Research Group, University of Debrecen, H-4032 Debrecen, Hungary
| | - Ferenc Erdődi
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
- MTA-DE Cell Biology and Signalling Research Group, University of Debrecen, H-4032 Debrecen, Hungary
- Correspondence: (F.E.); (A.K.); Tel.: +36-52-421345 (F.E. & A.K.)
| | - Andrea Kiss
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
- Correspondence: (F.E.); (A.K.); Tel.: +36-52-421345 (F.E. & A.K.)
| |
Collapse
|
12
|
Xu Y, Payne K, Pham LHG, Eunwoo P, Xiao J, Chi D, Lyu J, Campion R, Wasnik S, Jeong IS, Tang X, Baylink DJ, Chen CS, Reeves M, Akhtari M, Mirshahidi S, Marcucci G, Cao H. A novel vitamin D gene therapy for acute myeloid leukemia. Transl Oncol 2020; 13:100869. [PMID: 32956997 PMCID: PMC7509076 DOI: 10.1016/j.tranon.2020.100869] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 11/28/2022] Open
Abstract
Current treatment approaches for older adult patients with acute myeloid leukemia (AML) are often toxic and lack efficacy. Active vitamin D3 (1,25(OH)2D3) has been shown to induce myeloid blast differentiation but at concentrations that have resulted in unacceptable, off-target hypercalcemia in clinical trials. In our study, we found that the combination of 1,25(OH)2D3 and the hypomethylating agent (HMA) 5-Azacytidine (AZA) enhanced cytotoxicity and differentiation, and inhibited proliferation of several AML cell lines (MOLM-14, HL60) and primary AML patient samples. This observation was corroborated by our RNA sequence analysis data in which VDR, CD14, and BAX expression were increased, and FLT-3, PIM1 and Bcl-2 expression were decreased. To address the hypercalcemia issue, we genetically engineered MOLM-14 cells to constantly express CYP27B1 (the VD3 activating enzyme, 1-α-hydroxylase-25(OH)D3) through lentiviral transduction procedures. Subsequently, we used these cells as vehicles to deliver the CYP27B1 enzyme to the bone marrow of AML mice. We observed that AML mice with CYP27B1 treatment had longer overall survival compared to no treatment and displayed no significant change in calcium level.
Collapse
Affiliation(s)
- Yi Xu
- Department of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA, USA; Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Kimberly Payne
- Department of Basic Sciences, Division of Anatomy, Loma Linda University, Loma Linda, CA, USA
| | - Linh Hoang Gia Pham
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Park Eunwoo
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jeffrey Xiao
- Department of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA, USA; Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA, USA
| | - David Chi
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Justin Lyu
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Rosalia Campion
- Department of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA, USA
| | - Samiksha Wasnik
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Il Seok Jeong
- Department of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA, USA
| | - Xiaolei Tang
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA, USA; Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY, USA
| | - David Jeston Baylink
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Chien Shing Chen
- Department of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA, USA
| | - Mark Reeves
- Department of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA, USA
| | - Mojtaba Akhtari
- Department of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA, USA
| | - Saied Mirshahidi
- Loma Linda University Cancer Center Biospecimen Laboratory, Department of Medicine & Basic Sciences, Loma Linda, CA, USA
| | - Guido Marcucci
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Huynh Cao
- Department of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA, USA.
| |
Collapse
|
13
|
Integrating the Tumor Microenvironment into Cancer Therapy. Cancers (Basel) 2020; 12:cancers12061677. [PMID: 32599891 PMCID: PMC7352326 DOI: 10.3390/cancers12061677] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/11/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor progression is mediated by reciprocal interaction between tumor cells and their surrounding tumor microenvironment (TME), which among other factors encompasses the extracellular milieu, immune cells, fibroblasts, and the vascular system. However, the complexity of cancer goes beyond the local interaction of tumor cells with their microenvironment. We are on the path to understanding cancer from a systemic viewpoint where the host macroenvironment also plays a crucial role in determining tumor progression. Indeed, growing evidence is emerging on the impact of the gut microbiota, metabolism, biomechanics, and the neuroimmunological axis on cancer. Thus, external factors capable of influencing the entire body system, such as emotional stress, surgery, or psychosocial factors, must be taken into consideration for enhanced management and treatment of cancer patients. In this article, we review prognostic and predictive biomarkers, as well as their potential evaluation and quantitative analysis. Our overarching aim is to open up new fields of study and intervention possibilities, within the framework of an integral vision of cancer as a functional tissue with the capacity to respond to different non-cytotoxic factors, hormonal, immunological, and mechanical forces, and others inducing stroma and tumor reprogramming.
Collapse
|
14
|
Soto JR, Anthias C, Madrigal A, Snowden JA. Insights Into the Role of Vitamin D as a Biomarker in Stem Cell Transplantation. Front Immunol 2020; 11:966. [PMID: 32582151 PMCID: PMC7295104 DOI: 10.3389/fimmu.2020.00966] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 04/24/2020] [Indexed: 12/19/2022] Open
Abstract
Vitamin D was discovered 100 years ago and since then multiple studies have consistently proved its effect on bone health and mineral metabolism. Further research has also explored its so-called "non-classical" biological effects, encompassing immune regulation and control of cell proliferation and differentiation. Vitamin D downregulates pro-inflammatory immune cells and subsequently their cytokine production, while enhancing the anti-inflammatory subsets, thus mediating inflammation and fostering a more tolerogenic environment. Its biological action is exerted through the vitamin D receptor, a nuclear receptor that mediates gene transcription and is expressed in most cells from the innate and adaptive immunity. Owing to its immune-modulatory properties, its role in cancer pathophysiology, hematology disorders and stem cell transplantation has also been investigated. Vitamin D deficiency causes immune imbalance and cytokine dysregulation, contributing to some autoimmune diseases. In the hematopoietic stem cell transplant setting this could lead to complications such as acute and chronic graft-versus-host disease, ultimately impacting transplant outcomes. Other factors have also been linked to this, including specific polymorphisms of the vitamin D receptor in both stem cell donors and recipients. Nevertheless, studies thus far have shown conflicting results and the use of vitamin D or its receptor as biomarkers has not been validated yet, therefore there are no evidence-based consensus guidelines to guide clinicians in their day-to-day practice. To gain more insight in this topic, we have reviewed the existent literature and gathered the current evidence. This is an overview of the role of serum vitamin D and its receptor as biomarkers for clinical outcomes in patients undergoing hematopoietic stem cell transplantation. Further prospective studies with larger cohorts are warranted to validate the viability of using serum vitamin D, and its receptor, as biomarkers in potential stem cell donors and patients, to identify those at risk of post-transplant complications and enable early therapeutic interventions.
Collapse
Affiliation(s)
- Jose Ros Soto
- Anthony Nolan Research Institute, Royal Free Hospital and University College London, London, United Kingdom
| | - Chloe Anthias
- Anthony Nolan Research Institute, Royal Free Hospital and University College London, London, United Kingdom
- Department of Haemato-Oncology, The Royal Marsden Hospital, Sutton, United Kingdom
| | - Alejandro Madrigal
- Anthony Nolan Research Institute, Royal Free Hospital and University College London, London, United Kingdom
| | - John A. Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| |
Collapse
|
15
|
Carvalho J. Cell Reversal From a Differentiated to a Stem-Like State at Cancer Initiation. Front Oncol 2020; 10:541. [PMID: 32351900 PMCID: PMC7174973 DOI: 10.3389/fonc.2020.00541] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022] Open
Abstract
Even if the Somatic Mutation Theory of carcinogenesis explains many of the relevant experimental results in tumor origin and development, there are frequent events that are not justified, or are even contradictory to this widely accepted theory. A Cell Reversal Theory is presented, putting forward the hypothesis that cancer is originated by reversal of a differentiated cell into a non-differentiated stem-like state, by a change of its intrinsic epigenetic state, following a perturbation on the cell and/or its microenvironment. In the current proposal a cluster of cancer stem cells can be established, without the strict control mechanisms of a normal stem cell niche, and initiate a tumor. It is proposed that a reversal to a pluripotent state is at tumor origin and not tumor progress that prompts cell dedifferentiation. The uncontrolled proliferation of cancer stem cells causes a microenvironment disorganization, resulting in stressful conditions, like hypoxia and nutrient deprivation, which induces the genetic instability characteristic of a tumor; thus, in most cases, mutations are a consequence and not the direct cause of a tumor. It is also proposed that metastases result from dedifferentiation signaling dispersion instead of cell migration. However, conceivably, once the microenvironment is normalized, the stem cell-like state can differentiate back to a mature cell state and loose its oncogenic capacity. Therefore, this can be a reversible condition, suggesting important therapeutic opportunities.
Collapse
Affiliation(s)
- João Carvalho
- CFisUC, Department of Physics, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
16
|
Bizzarri M, Giuliani A, Cucina A, Minini M. Redifferentiation therapeutic strategies in cancer. Drug Discov Today 2020; 25:731-738. [PMID: 32027971 DOI: 10.1016/j.drudis.2020.01.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/10/2020] [Accepted: 01/28/2020] [Indexed: 12/21/2022]
Abstract
The widely recognized problems of pharmacological strategies based on killing cancer cells demand a rethink of therapeutic approaches. Tumor reversion strategies that aim to shift cancer cells to a healthy differentiated state are a promising alternative. Although many studies have firmly demonstrated the possibility of reverting cancer to a normal differentiated state, we are still unable (with the exception of retinoic acid in a form of leukemia) to revert cancer cells to a stable differentiated healthy state. Here, we review the main biological bases of redifferentiation strategies and provide a description of the most promising research avenues.
Collapse
Affiliation(s)
- Mariano Bizzarri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; Systems Biology Group Lab, Sapienza University, Rome, Italy.
| | | | - Alessandra Cucina
- Department of Surgery 'Pietro Valdoni', Sapienza University of Rome, 00161 Rome, Italy; Azienda Policlinico Umberto I, 00161 Rome, Italy
| | - Mirko Minini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; Department of Surgery 'Pietro Valdoni', Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
17
|
Nurminen V, Neme A, Seuter S, Carlberg C. Modulation of vitamin D signaling by the pioneer factor CEBPA. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1862:96-106. [PMID: 30550771 DOI: 10.1016/j.bbagrm.2018.12.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/06/2018] [Accepted: 12/06/2018] [Indexed: 01/31/2023]
Abstract
The myeloid master regulator CCAAT enhancer-binding protein alpha (CEBPA) is known as a pioneer factor. In this study, we report the CEBPA cistrome of THP-1 human monocytes after stimulation with the vitamin D receptor (VDR) ligand 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) for 2, 8 and 24 h. About a third of the genomic VDR binding sites co-located with those of CEBPA. In parallel, the binding strength of 5% of the CEBPA cistrome, i.e. some 1500 sites, is significantly (p < 0.001) affected by 1,25(OH)2D3. Transcriptome-wide analysis after CEBPA silencing indicated that the pioneer factor enhances both the basal expression and ligand inducibility of 70 vitamin D target genes largely involved in lipid signaling and metabolism. In contrast, CEBPA suppresses 82 vitamin D target genes many of which are related to the modulation of T cell activity by monocytes. The inducibility of the promoter-specific histone marker H3K4me3 distinguishes the former class of genes from the latter. Moreover, prominent occupancy of the myeloid pioneer factor PU.1 on 1,25(OH)2D3-sensitive CEBPA enhancers mechanistically explains the dichotomy of vitamin D target genes. In conclusion, CEBPA supports vitamin D signaling concerning actions of the innate immune system, but uses the antagonism with PU.1 for suppressing possible overreactions of adaptive immunity.
Collapse
Affiliation(s)
- Veijo Nurminen
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Antonio Neme
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Sabine Seuter
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Carsten Carlberg
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland.
| |
Collapse
|
18
|
Medrano M, Carrillo-Cruz E, Montero I, Perez-Simon JA. Vitamin D: Effect on Haematopoiesis and Immune System and Clinical Applications. Int J Mol Sci 2018; 19:ijms19092663. [PMID: 30205552 PMCID: PMC6164750 DOI: 10.3390/ijms19092663] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 08/31/2018] [Accepted: 09/04/2018] [Indexed: 12/19/2022] Open
Abstract
Vitamin D is a steroid-like hormone which acts by binding to vitamin D receptor (VDR). It plays a main role in the calcium homeostasis and metabolism. In addition, vitamin D display other important effects called “non-classical actions.” Among them, vitamin D regulates immune cells function and hematopoietic cells differentiation and proliferation. Based on these effects, it is currently being evaluated for the treatment of hematologic malignancies. In addition, vitamin D levels have been correlated with patients’ outcome after allogeneic stem cell transplantation, where it might regulate immune response and, accordingly, might influence the risk of graft-versus-host disease. Here, we present recent advances regarding its clinical applications both in the treatment of hematologic malignancies and in the transplant setting.
Collapse
Affiliation(s)
- Mayte Medrano
- Department of Hematology, University Hospital Virgen del Rocio, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Universidad de Sevilla, 41013 Sevilla, Spain.
| | - Estrella Carrillo-Cruz
- Department of Hematology, University Hospital Virgen del Rocio, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Universidad de Sevilla, 41013 Sevilla, Spain.
| | - Isabel Montero
- Department of Hematology, University Hospital Virgen del Rocio, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Universidad de Sevilla, 41013 Sevilla, Spain.
| | - Jose A Perez-Simon
- Department of Hematology, University Hospital Virgen del Rocio, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Universidad de Sevilla, 41013 Sevilla, Spain.
| |
Collapse
|
19
|
Abstract
The concept of differentiation therapy emerged from the fact that hormones or cytokines may promote differentiation ex vivo, thereby irreversibly changing the phenotype of cancer cells. Its hallmark success has been the treatment of acute promyelocytic leukaemia (APL), a condition that is now highly curable by the combination of retinoic acid (RA) and arsenic. Recently, drugs that trigger differentiation in a variety of primary tumour cells have been identified, suggesting that they are clinically useful. This Opinion article analyses the basis for the clinical successes of RA or arsenic in APL by assessing the respective roles of terminal maturation and loss of self-renewal. By reviewing other successful examples of drug-induced tumour cell differentiation, novel approaches to transform differentiating drugs into more efficient therapies are proposed.
Collapse
Affiliation(s)
- Hugues de Thé
- Collège de France, PSL Research University, 75005 Paris; Université Paris Diderot, Sorbonne Paris Cité (INSERM UMR 944, Equipe Labellisée par la Ligue Nationale contre le Cancer; CNRS UMR 7212), Institut Universitaire d'Hématologie, 75010 Paris; and Assistance Publique/Hôpitaux de Paris, Oncologie Moléculaire, Hôpital St Louis, 75010 Paris, France
| |
Collapse
|
20
|
Cortes M, Chen MJ, Stachura DL, Liu SY, Kwan W, Wright F, Vo LT, Theodore LN, Esain V, Frost IM, Schlaeger TM, Goessling W, Daley GQ, North TE. Developmental Vitamin D Availability Impacts Hematopoietic Stem Cell Production. Cell Rep 2017; 17:458-468. [PMID: 27705794 PMCID: PMC5338633 DOI: 10.1016/j.celrep.2016.09.012] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 07/18/2016] [Accepted: 09/02/2016] [Indexed: 01/01/2023] Open
Abstract
Vitamin D insufficiency is a worldwide epidemic affecting billions of individuals, including pregnant women and children. Despite its high incidence, the impact of active vitamin D3 (1,25(OH)D3) on embryonic development beyond osteo-regulation remains largely undefined. Here, we demonstrate that 1,25(OH)D3 availability modulates zebrafish hematopoietic stem and progenitor cell (HSPC) production. Loss of Cyp27b1-mediated biosynthesis or vitamin D receptor (VDR) function by gene knockdown resulted in significantly reduced runx1 expression and Flk1+cMyb+ HSPC numbers. Selective modulation in vivo and in vitro in zebrafish indicated that vitamin D3 acts directly on HSPCs, independent of calcium regulation, to increase proliferation. Notably, ex vivo treatment of human HSPCs with 1,25(OH)D3 also enhanced hematopoietic colony numbers, illustrating conservation across species. Finally, gene expression and epistasis analysis indicated that CXCL8 (IL-8) was a functional target of vitamin D3-mediated HSPC regulation. Together, these findings highlight the relevance of developmental 1,25(OH)D3 availability for definitive hematopoiesis and suggest potential therapeutic utility in HSPC expansion.
Collapse
Affiliation(s)
- Mauricio Cortes
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | - David L Stachura
- Department of Biological Sciences, California State University, Chico, Chico, CA 95929, USA
| | - Sarah Y Liu
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wanda Kwan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Francis Wright
- Department of Biological Sciences, California State University, Chico, Chico, CA 95929, USA
| | - Linda T Vo
- Boston Children's Hospital, Boston, MA 02115, USA
| | - Lindsay N Theodore
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Virginie Esain
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Isaura M Frost
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | - Wolfram Goessling
- Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - George Q Daley
- Boston Children's Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Trista E North
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
21
|
Dimitrov V, White JH. Vitamin D signaling in intestinal innate immunity and homeostasis. Mol Cell Endocrinol 2017; 453:68-78. [PMID: 28412519 DOI: 10.1016/j.mce.2017.04.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/10/2017] [Accepted: 04/10/2017] [Indexed: 12/14/2022]
Abstract
The lumen of the gut hosts a plethora of microorganisms that participate in food assimilation, inactivation of harmful particles and in vitamin synthesis. On the other hand, enteric flora, a number of food antigens, and toxins are capable of triggering immune responses causing inflammation, which, when unresolved, may lead to chronic conditions such as inflammatory bowel disease (IBD). It is important, therefore, to contain the gut bacteria within the lumen, control microbial load and composition, as well as ensure adequate innate and adaptive immune responses to pathogenic threats. There is growing evidence that vitamin D signaling has impacts on all these aspects of intestinal physiology, contributing to healthy enteric homeostasis. VD was first discovered as the curative agent for nutritional rickets, and its classical actions are associated with calcium absorption and bone health. However, vitamin D exhibits a number of extra-skeletal effects, particularly in innate immunity. Notably, it stimulates production of pattern recognition receptors, anti-microbial peptides, and cytokines, which are at the forefront of innate immune responses. They play a role in sensing the microbiota, in preventing excessive bacterial overgrowth, and complement the actions of vitamin D signaling in enhancing intestinal barrier function. Vitamin D also favours tolerogenic rather than inflammogenic T cell differentiation and function. Compromised innate immune function and overactive adaptive immunity, as well as defective intestinal barrier function, have been associated with IBD. Importantly, observational and intervention studies support a beneficial role of vitamin D supplementation in patients with Crohn's disease, a form of IBD. This review summarizes the effects of vitamin D signaling on barrier integrity and innate and adaptive immunity in the gut, as well as on microbial load and composition. Collectively, studies to date reveal that vitamin D signaling has widespread effects on gut homeostasis, and provide a mechanistic basis for potential therapeutic benefit of vitamin D supplementation in IBD.
Collapse
Affiliation(s)
- Vassil Dimitrov
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - John H White
- Department of Physiology, McGill University, Montreal, Quebec, Canada; Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
22
|
Zou H, Zhu XX, Zhang GB, Ma Y, Wu Y, Huang DS. Silibinin: an old drug for hematological disorders. Oncotarget 2017; 8:89307-89314. [PMID: 29179521 PMCID: PMC5687691 DOI: 10.18632/oncotarget.19153] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/27/2017] [Indexed: 12/16/2022] Open
Abstract
Introduction Silibinin (silybin), a non-toxic natural polyphenolic flavonoid, is the principal and the most biologically active component of silymarin. It is efficient in the treatment of acute and chronic liver disorders caused by toxins, drug, alcohol, hepatitis, and gall bladder disorders. Further, in our previous studies, we explored the anti-cancer efficacy in common cancers, such as lung, prostatic, colon, breast, bladder, as well as, hepatocellular carcinoma. Interestingly, silibinin is still not solely limited to the treatment of these diseases. Recent research endeavors suggest that silibinin may function diversely and serve as a novel therapy for hematological disorders. Areas covered It discovered several interesting viewpoints in the widely studied mechanisms of silibinin in the hematological disorders. Expert commentary In this report, we review the up-to-date findings of more potency roles of silibinin in β-thalassemia (β-TM), acute myeloid leukemia (AML), anaplastic large cell lymphoma (ALCL) and multiple myelomas (MM) therapy and attempt to clarify the mechanisms underlying its effects. There are two viewpoints: First, The functional mechanisms of silibinin in AML cells via regulating cell differentiation to exert anti-cancer effect; Second, combination treatment strategy may be a good choice.
Collapse
Affiliation(s)
- Hai Zou
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou 310000, China.,People's Hospital of Hangzhou Medical College, Hangzhou 310000, China
| | - Xing-Xing Zhu
- Department of Nephrology, Zhejiang Provincial People's Hospital, Hangzhou 310000, China.,People's Hospital of Hangzhou Medical College, Hangzhou 310000, China
| | - Guo-Bing Zhang
- Department of Pharmacy, Zhejiang Provincial People's Hospital, Hangzhou 310000, China.,People's Hospital of Hangzhou Medical College, Hangzhou 310000, China
| | - Yuan Ma
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou 310000, China.,People's Hospital of Hangzhou Medical College, Hangzhou 310000, China
| | - Yi Wu
- Department of Hematology, Zhejiang Provincial People's Hospital, Hangzhou 310000, China.,People's Hospital of Hangzhou Medical College, Hangzhou 310000, China
| | - Dong-Sheng Huang
- Department of Hepatobiliary Surgery, Zhejiang Provincial People's Hospital, Hangzhou 310000, China.,People's Hospital of Hangzhou Medical College, Hangzhou 310000, China
| |
Collapse
|
23
|
Application of vitamin D and vitamin D analogs in acute myelogenous leukemia. Exp Hematol 2017; 50:1-12. [DOI: 10.1016/j.exphem.2017.01.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/16/2017] [Accepted: 01/26/2017] [Indexed: 12/18/2022]
|
24
|
Identification of H7 as a novel peroxiredoxin I inhibitor to induce differentiation of leukemia cells. Oncotarget 2016; 7:3873-83. [PMID: 26716647 PMCID: PMC4826176 DOI: 10.18632/oncotarget.6763] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 11/28/2015] [Indexed: 01/02/2023] Open
Abstract
Identifying novel targets to enhance leukemia-cell differentiation is an urgent requirment. We have recently proposed that inhibiting the antioxidant enzyme peroxiredoxin I (Prdx I) may induce leukemia-cell differentiation. However, this concept remains to be confirmed. In this work, we identified H7 as a novel Prdx I inhibitor through virtual screening, in vitro activity assay, and surface plasmon resonance assay. Cellular thermal shift assay showed that H7 directly bound to Prdx I but not to Prdxs II–V in cells. H7 treatment also increased reactive oxygen species (ROS) level and cell differentiation in leukemia cells, as reflected by the upregulation of the cell surface differentiation marker CD11b/CD14 and the morphological maturation of cells. The differentiation-induction effect of H7 was further observed in some non-acute promyelocytic leukemia (APL) and primary leukemia cells apart from APL NB4 cells. Moreover, the ROS scavenger N-acetyl cysteine significantly reversed the H7-induced cell differentiation. We demonstrated as well that H7-induced cell differentiation was associated with the activation of the ROS-Erk1/2-C/EBPβ axis. Finally, we showed H7 treatment induced cell differentiation in an APL mouse model. All of these data confirmed that Prdx I was novel target for inducing leukemia-cell differentiation and that H7 was a novel lead compound for optimizing Prdx I inhibition.
Collapse
|
25
|
García-Ramírez I, Martín-Lorenzo A, González-Herrero I, Rodriguez-Hernández G, Vicente-Dueñas C, Sánchez-García I. Could Vitamin D Analogues Be Used to Target Leukemia Stem Cells? Int J Mol Sci 2016; 17:ijms17060889. [PMID: 27275819 PMCID: PMC4926423 DOI: 10.3390/ijms17060889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/23/2016] [Accepted: 05/31/2016] [Indexed: 11/17/2022] Open
Abstract
Leukemic stem cells (LSCs) are defined as cells that possess the ability to self-renew and give rise to the differentiated cancer cells that comprise the tumor. These LSCs seem to show chemo-resistance and radio-resistance leading to the failure of conventional cancer therapies. Current therapies are directed at the fast growing tumor mass leaving the LSC fraction untouched. Eliminating LSCs, the root of cancer origin and recurrence, is considered to be a hopeful approach to improve survival or even to cure cancer patients. In order to achieve this, the characterization of LSCs is a prerequisite in order to develop LSC-based therapies to eliminate them. Here we review if vitamin D analogues may allow an avenue to target the LSCs.
Collapse
Affiliation(s)
- Idoia García-Ramírez
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Campus M. de Unamuno s/n, Salamanca 37007, Spain.
- Cancer Research Area, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain.
| | - Alberto Martín-Lorenzo
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Campus M. de Unamuno s/n, Salamanca 37007, Spain.
- Cancer Research Area, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain.
| | - Inés González-Herrero
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Campus M. de Unamuno s/n, Salamanca 37007, Spain.
- Cancer Research Area, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain.
| | - Guillermo Rodriguez-Hernández
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Campus M. de Unamuno s/n, Salamanca 37007, Spain.
- Cancer Research Area, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain.
| | - Carolina Vicente-Dueñas
- Cancer Research Area, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain.
| | - Isidro Sánchez-García
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Campus M. de Unamuno s/n, Salamanca 37007, Spain.
- Cancer Research Area, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain.
| |
Collapse
|
26
|
Marcinkowska E, Wallace GR, Brown G. The Use of 1α,25-Dihydroxyvitamin D₃ as an Anticancer Agent. Int J Mol Sci 2016; 17:E729. [PMID: 27187375 PMCID: PMC4881551 DOI: 10.3390/ijms17050729] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 04/22/2016] [Accepted: 05/10/2016] [Indexed: 12/12/2022] Open
Abstract
The notion that vitamin D can influence the incidence of cancer arose from epidemiological studies. The major source of vitamin D in the organism is skin production upon exposure to ultra violet-B. The very first observation of an inverse correlation between exposure of individuals to the sun and the likelihood of cancer was reported as early as 1941. In 1980, Garland and Garland hypothesised, from findings from epidemiological studies of patients in the US with colon cancer, that vitamin D produced in response to sun exposure is protective against cancer as opposed to sunlight per se. Later studies revealed inverse correlations between sun exposure and the occurrence of prostate and breast cancers. These observations prompted laboratory investigation of whether or not vitamin D had an effect on cancer cells. Vitamin D is not active against cancer cells, but the most active metabolite 1α,25-dihydroxyvitamin D₃ (1,25D) has profound biological effects. Here, we review the anticancer action of 1,25D, clinical trials of 1,25D to date and the prospects of the future therapeutic use of new and low calcaemic analogues.
Collapse
Affiliation(s)
- Ewa Marcinkowska
- Laboratory of Protein Biochemistry, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Graham R Wallace
- Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Geoffrey Brown
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
27
|
Molaeipour Z, Shamsasanjan K, Movassaghpour AA, Akbarzadehlaleh P, Sabaghi F, Saleh M. The Effect of Bone Marrow Mesenchymal Stem Cells on Vitamin D3 Induced Monocytic Differentiation of U937 Cells. Adv Pharm Bull 2016; 6:23-9. [PMID: 27123414 DOI: 10.15171/apb.2016.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/27/2015] [Accepted: 01/07/2016] [Indexed: 12/30/2022] Open
Abstract
PURPOSE Mesenchymal stem cells (MSCs) are key components of the hematopoietic stem cells (HSCs) niche. They control the process of hematopoiesis by secreting regulatory cytokines, growth factors and expression of important cell adhesion molecules for cell-tocell interactions. In this research, we have investigated the effect of bone marrow derived MSCs on monocytic differentiation of U937 cells line. METHODS U937 cells were cultured in both direct co-culture with MSCs and MSCs conditioned medium (C.M) driven. This study used 1,25-dihydroxyvitamin D3(VitD3) as inductor of monocytic differentiation and U937 cells treated with VitD3 morphology was examined by Wright Giemsa staining. CD14 monocytic differentiation marker was measured by flow cytometry and monocytic gene expression was assessed by real time polymerase chain reaction (RT PCR). RESULTS The results of flow cytometric analysis showed that CD14 expression of U937 increased. The higher effect of MSCs co-culture on CD14 expression in U937 cells was observed, compared to the conditioned medium. Among ten monocytic related genes which were screened that was observed increase in 5 genes in which CXCR4 and CSF2RA showed significant increase. CONCLUSION The results obtained show that MSCs have supportive effect on the monocytic differentiation of U937 cells. However, a distinct mechanism of that remains unclear.
Collapse
Affiliation(s)
- Zahra Molaeipour
- Hematology Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasanjan
- Hematology Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Parvin Akbarzadehlaleh
- Pharmaceutical Biotechnology Department, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sabaghi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mahshid Saleh
- Hematology Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
28
|
Molaeipour Z, Shamsasanjan K, Movassaghpour AA, Akbarzadehlaleh P, Sabaghi F, Saleh M. The Effect of Bone Marrow Mesenchymal Stem Cells on Vitamin D3 Induced Monocytic Differentiation of U937 Cells. Adv Pharm Bull 2016; 6:23-29. [PMID: 27123414 PMCID: PMC4845548 DOI: 10.15171/apb.2016.05] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/27/2015] [Accepted: 01/07/2016] [Indexed: 08/05/2023] Open
Abstract
PURPOSE Mesenchymal stem cells (MSCs) are key components of the hematopoietic stem cells (HSCs) niche. They control the process of hematopoiesis by secreting regulatory cytokines, growth factors and expression of important cell adhesion molecules for cell-tocell interactions. In this research, we have investigated the effect of bone marrow derived MSCs on monocytic differentiation of U937 cells line. METHODS U937 cells were cultured in both direct co-culture with MSCs and MSCs conditioned medium (C.M) driven. This study used 1,25-dihydroxyvitamin D3(VitD3) as inductor of monocytic differentiation and U937 cells treated with VitD3 morphology was examined by Wright Giemsa staining. CD14 monocytic differentiation marker was measured by flow cytometry and monocytic gene expression was assessed by real time polymerase chain reaction (RT PCR). RESULTS The results of flow cytometric analysis showed that CD14 expression of U937 increased. The higher effect of MSCs co-culture on CD14 expression in U937 cells was observed, compared to the conditioned medium. Among ten monocytic related genes which were screened that was observed increase in 5 genes in which CXCR4 and CSF2RA showed significant increase. CONCLUSION The results obtained show that MSCs have supportive effect on the monocytic differentiation of U937 cells. However, a distinct mechanism of that remains unclear.
Collapse
Affiliation(s)
- Zahra Molaeipour
- Hematology Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasanjan
- Hematology Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Parvin Akbarzadehlaleh
- Pharmaceutical Biotechnology Department, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sabaghi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mahshid Saleh
- Hematology Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
29
|
Radujkovic A, Schnitzler P, Ho AD, Dreger P, Luft T. Low serum vitamin D levels are associated with shorter survival after first-line azacitidine treatment in patients with myelodysplastic syndrome and secondary oligoblastic acute myeloid leukemia. Clin Nutr 2016; 36:542-551. [PMID: 26899917 DOI: 10.1016/j.clnu.2016.01.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 01/21/2016] [Accepted: 01/25/2016] [Indexed: 10/22/2022]
Abstract
BACKGROUND & AIMS Azacitidine (AZA) therapy has become the recommended first-line treatment for patients with high-risk myelodysplastic syndromes (MDS) and oligoblastic (<30% bone marrow blasts) acute myeloid leukemia (AML). However, improvement of the efficacy of AZA treatment remains a challenge. We retrospectively tested the hypothesis that VitD levels (25-hydroxyvitamin D3) prior to start of first-line AZA therapy are predictive of overall survival (OS) in patients diagnosed with MDS and secondary oligoblastic AML. Furthermore, the antiproliferative effects of AZA in combination with 25-hydroxyvitamin D3 and 1α,25-dihydroxyvitamin D3 were investigated in vitro. METHODS A total of 58 patients treated at our center between 2006 and 2014 were analyzed. Serum levels of VitD were quantified using a standard, commercially available 25-hydroxyvitamin D3 chemiluminescent immunoassay. Effects on cell proliferation were assessed using tetrazolium-based MTT assays. RESULTS Median serum VitD level prior to AZA treatment was 32.8 nM (range 11.0-101.5 nM). Patient, disease and treatment characteristics did not differ significantly between the low (≤32.8 nM; n = 29) and high (>32.8 nM; n = 29) VitD group. Estimated probability of 2-year OS in the low versus high VitD group was 14% versus 40% (P < 0.05). In multivariable analysis with OS as endpoint, adverse cytogenetics (HR 2.66, P = 0.03) and VitD (per 10 nM decrease, HR 1.68, P = 0.02) were independent predictors of worse survival. In-vitro treatment of myeloid cell lines with AZA in combination with VitD produced synergistic and additive antiproliferative effects. Addition of nanomolar VitD concentrations to AZA resulted in potentiation of AZA activity. Conversely, combination with the VitD antagonist TEI-9647 resulted in inhibition of AZA activity. CONCLUSIONS Our study suggests that higher VitD levels were associated with a survival advantage following first-line AZA therapy. Enhanced cytotoxic effects upon combination treatment may contribute to the observed clinical effects. VitD repletion/supplementation during AZA treatment should be explored.
Collapse
Affiliation(s)
- Aleksandar Radujkovic
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany.
| | - Paul Schnitzler
- Department of Infectious Diseases, Virology, Heidelberg, Germany
| | - Anthony D Ho
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Luft
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
30
|
Ong E, Szedlak A, Kang Y, Smith P, Smith N, McBride M, Finlay D, Vuori K, Mason J, Ball ED, Piermarocchi C, Paternostro G. A scalable method for molecular network reconstruction identifies properties of targets and mutations in acute myeloid leukemia. J Comput Biol 2016; 22:266-88. [PMID: 25844667 DOI: 10.1089/cmb.2014.0297] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A key aim of systems biology is the reconstruction of molecular networks. We do not yet, however, have networks that integrate information from all datasets available for a particular clinical condition. This is in part due to the limited scalability, in terms of required computational time and power, of existing algorithms. Network reconstruction methods should also be scalable in the sense of allowing scientists from different backgrounds to efficiently integrate additional data. We present a network model of acute myeloid leukemia (AML). In the current version (AML 2.1), we have used gene expression data (both microarray and RNA-seq) from 5 different studies comprising a total of 771 AML samples and a protein-protein interactions dataset. Our scalable network reconstruction method is in part based on the well-known property of gene expression correlation among interacting molecules. The difficulty of distinguishing between direct and indirect interactions is addressed by optimizing the coefficient of variation of gene expression, using a validated gold-standard dataset of direct interactions. Computational time is much reduced compared to other network reconstruction methods. A key feature is the study of the reproducibility of interactions found in independent clinical datasets. An analysis of the most significant clusters, and of the network properties (intraset efficiency, degree, betweenness centrality, and PageRank) of common AML mutations demonstrated the biological significance of the network. A statistical analysis of the response of blast cells from 11 AML patients to a library of kinase inhibitors provided an experimental validation of the network. A combination of network and experimental data identified CDK1, CDK2, CDK4, and CDK6 and other kinases as potential therapeutic targets in AML.
Collapse
|
31
|
Zhang H, Qian PY, Ravasi T. Selective phosphorylation during early macrophage differentiation. Proteomics 2015; 15:3731-43. [PMID: 26307563 DOI: 10.1002/pmic.201400511] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 07/08/2015] [Accepted: 08/19/2015] [Indexed: 12/27/2022]
Abstract
The differentiation of macrophages from monocytes is a tightly controlled and complex biological process. Although numerous studies have been conducted using biochemical approaches or global gene/protein profiling, the mechanisms of the early stages of differentiation remain unclear. Here we used SILAC-based quantitative proteomics approach to perform temporal phosphoproteome profiling of early macrophage differentiation. We identified a large set of phosphoproteins and grouped them as PMA-regulated and non-regulated phosphoproteins in the early stages of differentiation. Further analysis of the PMA-regulated phosphoproteins revealed that transcriptional suppression, cytoskeletal reorganization and cell adhesion were among the most significantly activated pathways. Some key involved regulators of these pathways are mTOR, MYB, STAT1 and CTNNB. Moreover, we were able to classify the roles and activities of several transcriptional factors during different differentiation stages and found that E2F is likely to be an important regulator during the relatively late stages of differentiation. This study provides the first comprehensive picture of the dynamic phosphoproteome during myeloid cells differentiation, and identifies potential molecular targets in leukemic cells.
Collapse
Affiliation(s)
- Huoming Zhang
- Division of Biological and Environmental Sciences & Engineering, Division of Applied Mathematics and Computer Sciences, King Abdullah University of Science & Technology, Thuwal, Kingdom of Saudi Arabia.,Bioscience Core Laboratory, King Abdullah University of Science and Technology, Thuwal, Kingdom of Saudi Arabia
| | - Pei-Yuan Qian
- School of Science, Hong Kong University of Science and Technology, Hong Kong, P. R. China
| | - Timothy Ravasi
- Division of Biological and Environmental Sciences & Engineering, Division of Applied Mathematics and Computer Sciences, King Abdullah University of Science & Technology, Thuwal, Kingdom of Saudi Arabia
| |
Collapse
|
32
|
Paolillo R, Spinello I, Quaranta MT, Pasquini L, Pelosi E, Lo Coco F, Testa U, Labbaye C. Human TM9SF4 Is a New Gene Down-Regulated by Hypoxia and Involved in Cell Adhesion of Leukemic Cells. PLoS One 2015; 10:e0126968. [PMID: 25961573 PMCID: PMC4427288 DOI: 10.1371/journal.pone.0126968] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/09/2015] [Indexed: 12/19/2022] Open
Abstract
Background The transmembrane 9 superfamily protein member 4, TM9SF4, belongs to the TM9SF family of proteins highly conserved through evolution. TM9SF4 homologs, previously identified in many different species, were mainly involved in cellular adhesion, innate immunity and phagocytosis. In human, the function and biological significance of TM9SF4 are currently under investigation. However, TM9SF4 was found overexpressed in human metastatic melanoma and in a small subset of acute myeloid leukemia (AMLs) and myelodysplastic syndromes, consistent with an oncogenic function of this gene. Purpose and Results In this study, we first analyzed the expression and regulation of TM9SF4 in normal and leukemic cells and identified TM9SF4 as a gene highly expressed in human quiescent CD34+ hematopoietic progenitor cells (HPCs), regulated during monocytic and granulocytic differentiation of HPCs, both lineages giving rise to mature myeloid cells involved in adhesion, phagocytosis and immunity. Then, we found that TM9SF4 is markedly overexpressed in leukemic cells and in AMLs, particularly in M2, M3 and M4 AMLs (i.e., in AMLs characterized by the presence of a more or less differentiated granulocytic progeny), as compared to normal CD34+ HPCs. Proliferation and differentiation of HPCs occurs in hypoxia, a physiological condition in bone marrow, but also a crucial component of cancer microenvironment. Here, we investigated the impact of hypoxia on TM9SF4 expression in leukemic cells and identified TM9SF4 as a direct target of HIF-1α, downregulated in these cells by hypoxia. Then, we found that the hypoxia-mediated downregulation of TM9SF4 expression is associated with a decrease of cell adhesion of leukemic cells to fibronectin, thus demonstrating that human TM9SF4 is a new molecule involved in leukemic cell adhesion. Conclusions Altogether, our study reports for the first time the expression of TM9SF4 at the level of normal and leukemic hematopoietic cells and its marked expression at the level of AMLs displaying granulocytic differentiation.
Collapse
MESH Headings
- Apoptosis/drug effects
- Bone Marrow Cells/drug effects
- Bone Marrow Cells/metabolism
- Bone Marrow Cells/pathology
- Cell Adhesion/drug effects
- Cell Cycle/drug effects
- Cell Differentiation/drug effects
- Cell Hypoxia
- Cell Proliferation/drug effects
- Cloning, Molecular
- Fibronectins/metabolism
- Gene Expression Regulation, Leukemic
- Granulocytes/drug effects
- Granulocytes/metabolism
- Granulocytes/pathology
- HEK293 Cells
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Monocytes/drug effects
- Monocytes/metabolism
- Monocytes/pathology
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Oxygen/pharmacology
- Primary Cell Culture
- Promoter Regions, Genetic
- Signal Transduction
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Rosa Paolillo
- Department of Hematology, Oncology and Molecular Medicine of Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Isabella Spinello
- Department of Hematology, Oncology and Molecular Medicine of Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Maria Teresa Quaranta
- Department of Hematology, Oncology and Molecular Medicine of Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Luca Pasquini
- Department of Hematology, Oncology and Molecular Medicine of Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine of Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Francesco Lo Coco
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
- Fondazione Santa Lucia, Rome, Italy
| | - Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine of Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Catherine Labbaye
- Department of Hematology, Oncology and Molecular Medicine of Istituto Superiore di Sanità, 00161, Rome, Italy
- * E-mail:
| |
Collapse
|
33
|
Giammanco M, Di Majo D, La Guardia M, Aiello S, Crescimannno M, Flandina C, Tumminello FM, Leto G. Vitamin D in cancer chemoprevention. PHARMACEUTICAL BIOLOGY 2015; 53:1399-1434. [PMID: 25856702 DOI: 10.3109/13880209.2014.988274] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CONTEXT There is increasing evidence that Vitamin D (Vit D) and its metabolites, besides their well-known calcium-related functions, may also exert antiproliferative, pro-differentiating, and immune modulatory effects on tumor cells in vitro and may also delay tumor growth in vivo. OBJECTIVE The aim of this review is to provide fresh insight into the most recent advances on the role of Vit D and its analogues as chemopreventive drugs in cancer therapy. METHODS A systematic review of experimental and clinical studies on Vit D and cancer was undertaken by using the major electronic health database including ISI Web of Science, Medline, PubMed, Scopus and Google Scholar. RESULTS AND CONCLUSION Experimental and clinical observations suggest that Vit D and its analogues may be effective in preventing the malignant transformation and/or the progression of various types of human tumors including breast cancer, prostate cancer, colorectal cancer, and some hematological malignances. These findings suggest the possibility of the clinical use of these molecules as novel potential chemopreventive and anticancer agents.
Collapse
|
34
|
Teng CLJ, Han SM, Wu WC, Hsueh CM, Tsai JR, Hwang WL, Hsu SL. Mechanistic aspects of lauryl gallate-induced differentiation and apoptosis in human acute myeloid leukemia cells. Food Chem Toxicol 2014; 71:197-206. [DOI: 10.1016/j.fct.2014.06.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 06/12/2014] [Accepted: 06/23/2014] [Indexed: 11/29/2022]
|
35
|
Gemelli C, Martello A, Montanari M, Zanocco Marani T, Salsi V, Zappavigna V, Parenti S, Vignudelli T, Selmi T, Ferrari S, Grande A. The Orosomucoid 1 protein is involved in the vitamin D – mediated macrophage de-activation process. Exp Cell Res 2013; 319:3201-13. [DOI: 10.1016/j.yexcr.2013.08.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 08/06/2013] [Accepted: 08/14/2013] [Indexed: 12/20/2022]
|
36
|
Rahimian A, Soleimani M, Kaviani S, Aghaee-Bakhtiari SH, Atashi A, Arefian E, Nikougoftar M. Bypassing the maturation arrest in myeloid cell line U937 by over-expression of microRNA-424. Hematology 2013; 16:298-302. [DOI: 10.1179/102453311x13085644680140] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Affiliation(s)
- A Rahimian
- Department of HematologySchool of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Molecular Biology and Genetic EngineeringStem Cell Technology Research Center, Tehran, Iran
| | - M Soleimani
- Department of HematologySchool of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - S Kaviani
- Department of HematologySchool of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - S H Aghaee-Bakhtiari
- Department of Molecular Biology and Genetic EngineeringStem Cell Technology Research Center, Tehran, Iran
| | - A Atashi
- Department of Stem Cell Biology DepartmentStem Cell Technology Research Center, Tehran, Iran
| | - E Arefian
- Department of Molecular Biology and Genetic EngineeringStem Cell Technology Research Center, Tehran, Iran
| | - M Nikougoftar
- Research CenterIranian Blood Transfusion Organization (IBTO), Tehran, Iran
| |
Collapse
|
37
|
Rőszer T, Menéndez-Gutiérrez MP, Cedenilla M, Ricote M. Retinoid X receptors in macrophage biology. Trends Endocrinol Metab 2013; 24:460-8. [PMID: 23701753 DOI: 10.1016/j.tem.2013.04.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 04/19/2013] [Accepted: 04/23/2013] [Indexed: 01/07/2023]
Abstract
Retinoid X receptors (RXRs) form a distinct and unique subclass within the nuclear receptor (NR) superfamily of ligand-dependent transcription factors. RXRs regulate a plethora of genetic programs, including cell differentiation, the immune response, and lipid and glucose metabolism. Recent advances reveal that RXRs are important regulators of macrophages, key players in inflammatory and metabolic disorders. This review outlines the versatility of RXR action in the control of macrophage gene transcription through its heterodimerization with other NRs or through RXR homodimerization. We also highlight the potential of RXR-controlled transcriptional programs as targets for the treatment of pathologies associated with altered macrophage function, such as atherosclerosis, insulin resistance, autoimmunity, and neurodegeneration.
Collapse
Affiliation(s)
- Tamás Rőszer
- Cardiovascular Development and Repair Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | | | | | | |
Collapse
|
38
|
Lainey E, Wolfromm A, Sukkurwala AQ, Micol JB, Fenaux P, Galluzzi L, Kepp O, Kroemer G. EGFR inhibitors exacerbate differentiation and cell cycle arrest induced by retinoic acid and vitamin D3 in acute myeloid leukemia cells. Cell Cycle 2013; 12:2978-91. [PMID: 23974111 DOI: 10.4161/cc.26016] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
By means of an unbiased, automated fluorescence microscopy-based screen, we identified the epidermal growth factor receptor (EGFR) inhibitors erlotinib and gefitinib as potent enhancers of the differentiation of HL-60 acute myeloid leukemia (AML) cells exposed to suboptimal concentrations of vitamin A (all-trans retinoic acid, ATRA) or vitamin D (1α,25-hydroxycholecalciferol, VD). Erlotinib and gefitinib alone did not promote differentiation, yet stimulated the acquisition of morphological and biochemical maturation markers (including the expression of CD11b and CD14 as well as increased NADPH oxidase activity) when combined with either ATRA or VD. Moreover, the combination of erlotinib and ATRA or VD synergistically induced all the processes that are normally linked to terminal hematopoietic differentiation, namely, a delayed proliferation arrest in the G0/G1 phase of the cell cycle, cellular senescence, and apoptosis. Erlotinib potently inhibited the (auto)phosphorylation of mitogen-activated protein kinase 14 (MAPK14, best known as p38(MAPK)) and SRC family kinases (SFKs). If combined with the administration of ATRA or VD, the inhibition of p38(MAPK) or SFKs with specific pharmacological agents mimicked the pro-differentiation activity of erlotinib. These data were obtained with 2 distinct AML cell lines (HL-60 and MOLM-13 cells) and could be confirmed on primary leukemic blasts isolated from the circulation of AML patients. Altogether, these findings point to a new regimen for the treatment of AML, in which naturally occurring pro-differentiation agents (ATRA or VD) may be combined with EGFR inhibitors.
Collapse
Affiliation(s)
- Elodie Lainey
- INSERM; U848; Villejuif, France; Gustave Roussy; Villejuif, France; Université Paris Sud/Paris XI; Le Kremlin Bicêtre, France; Hôpital Robert Debré; AP-HP; Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Paubelle E, Zylbersztejn F, Alkhaeir S, Suarez F, Callens C, Dussiot M, Isnard F, Rubio MT, Damaj G, Gorin NC, Marolleau JP, Monteiro RC, Moura IC, Hermine O. Deferasirox and vitamin D improves overall survival in elderly patients with acute myeloid leukemia after demethylating agents failure. PLoS One 2013; 8:e65998. [PMID: 23840388 PMCID: PMC3688707 DOI: 10.1371/journal.pone.0065998] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 04/30/2013] [Indexed: 12/28/2022] Open
Abstract
The prognosis of acute myeloid leukemia (AML) in elderly (≥65 years) patients is poor and treatment remains non-consensual especially for those who are not eligible for intensive therapies. Our group has shown that in vitro the iron chelator deferasirox (DFX) synergizes with vitamin D (VD) to promote monocyte differentiation in primary AML cells. Herein, we present results from a retrospective case-control study in which the association of DFX (1-2 g/d) and 25-hydroxycholecalciferol (100,000 IU/week) (DFX/VD) was proposed to patients following demethylating agents failure. Median survival of patients treated with DFX/VD combination (n = 17) was significantly increased in comparison with matched patients receiving best supportive care (BSC) alone (n = 13) (10.4 versus 4 months respectively). In addition, the only factor associated to an increased overall survival in DFX/VD-treated patients was serum VD levels. We conclude that DFX/VD treatment correlated with increased overall survival of AML patients in this retrospective cohort of elderly patients.
Collapse
Affiliation(s)
- Etienne Paubelle
- CNRS UMR 8147, Université Paris Descartes, Faculté de Médecine, Hôpital Necker, Paris, France
- INSERM U699, Paris, France
- Insitut Hospitalo-Universitaire (IHU) Imagine, Université Sorbonne Paris cité, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
- Service d’Hématologie clinique, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Florence Zylbersztejn
- CNRS UMR 8147, Université Paris Descartes, Faculté de Médecine, Hôpital Necker, Paris, France
- INSERM U699, Paris, France
- Insitut Hospitalo-Universitaire (IHU) Imagine, Université Sorbonne Paris cité, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Sawsaneh Alkhaeir
- CNRS UMR 8147, Université Paris Descartes, Faculté de Médecine, Hôpital Necker, Paris, France
- INSERM U699, Paris, France
- Insitut Hospitalo-Universitaire (IHU) Imagine, Université Sorbonne Paris cité, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Felipe Suarez
- Insitut Hospitalo-Universitaire (IHU) Imagine, Université Sorbonne Paris cité, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
- Service d’Hématologie clinique, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Céline Callens
- CNRS UMR 8147, Université Paris Descartes, Faculté de Médecine, Hôpital Necker, Paris, France
| | - Michaël Dussiot
- CNRS UMR 8147, Université Paris Descartes, Faculté de Médecine, Hôpital Necker, Paris, France
- INSERM U699, Paris, France
- Insitut Hospitalo-Universitaire (IHU) Imagine, Université Sorbonne Paris cité, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
- Laboratoire d’excellence des Globules Rouges (GR-ex), Paris, France
| | - Françoise Isnard
- Department of Hematology, AP-HP Hôpital Saint Antoine, Université Pierre et Marie Curie, Paris, France
| | - Marie-Thérèse Rubio
- Department of Hematology, AP-HP Hôpital Saint Antoine, Université Pierre et Marie Curie, Paris, France
| | - Gandhi Damaj
- Centre Hospitalier Universitaire (CHU) Sud, Amiens, France
| | - Norbert-Claude Gorin
- Department of Hematology, AP-HP Hôpital Saint Antoine, Université Pierre et Marie Curie, Paris, France
| | | | - Renato C. Monteiro
- INSERM U699, Paris, France
- Service d’Immunologie, Hôpital Bichat, Assistance Publique Hôpitaux de Paris, Paris, France
- Faculté de Médecine and Université Denis Diderot Paris VII, Paris, France
| | - Ivan C. Moura
- INSERM U699, Paris, France
- Insitut Hospitalo-Universitaire (IHU) Imagine, Université Sorbonne Paris cité, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
- Faculté de Médecine and Université Denis Diderot Paris VII, Paris, France
| | - Olivier Hermine
- CNRS UMR 8147, Université Paris Descartes, Faculté de Médecine, Hôpital Necker, Paris, France
- Insitut Hospitalo-Universitaire (IHU) Imagine, Université Sorbonne Paris cité, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
- Service d’Hématologie clinique, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
- Laboratoire d’excellence des Globules Rouges (GR-ex), Paris, France
- * E-mail:
| |
Collapse
|
40
|
Hall AC, Juckett MB. The role of vitamin D in hematologic disease and stem cell transplantation. Nutrients 2013; 5:2206-21. [PMID: 23778150 PMCID: PMC3725501 DOI: 10.3390/nu5062206] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 06/03/2013] [Accepted: 06/08/2013] [Indexed: 11/16/2022] Open
Abstract
Vitamin D is a steroid hormone with a broad range of biological effects ranging from the classical role as a mediator of calcium and phosphate balance to cellular differentiation and immune modulation. These effects impact normal and dysfunctional hematopoietic and immune function, which may allow an avenue for improved treatment and support of patients suffering from hematologic disorders. In this review, we will summarize the role of vitamin D in normal hematopoiesis, discuss ways in which vitamin D may improve outcomes, and discuss a potential role of vitamin D for treating hematologic disorders and modulating the immune system to improve the outcome of allogeneic stem cell transplant.
Collapse
Affiliation(s)
- Aric C Hall
- Division of Hematology and Medical Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA.
| | | |
Collapse
|
41
|
1,25-Dihydroxyvitamin D3 promotes a sustained LPS-induced NF-κB-dependent expression of CD55 in human monocytic THP-1 cells. PLoS One 2012; 7:e49318. [PMID: 23152895 PMCID: PMC3495912 DOI: 10.1371/journal.pone.0049318] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 10/10/2012] [Indexed: 01/05/2023] Open
Abstract
The vitamin D3 system imposes immunosuppressive effects on monocytic cells, in part, by inhibiting NF-κB-dependent expression of proinflammatory mediators. CD55, a cell surface complement regulatory protein that promotes protective and anti-inflammatory properties, is reportedly an NF-κB target gene transiently induced in monocytic cells by the bacterial endotoxin LPS. CD55 is elevated on white cells in women experiencing preterm labor (a pathophysiology commonly associated with bacterial infection) and failure to maintain CD55 was associated with subsequent preterm delivery. We examined the influence of vitamin D3 signaling on LPS-induced expression of CD55 in human monocytic THP-1 cells using quantitative PCR, immunoblot, immunohistochemistry, and NF-κB activation pathway inhibitors. Non-NF-κB targets CD14 and CD11b, which modulate bacterial surveillance and eradication, respectively, were also examined. LPS produced a rapid transient 1.6-fold increase in CD55 mRNA. 1,25-D3 alone did not affect CD55 mRNA expression within the first 48 h. However, in 1,25-D3 pretreated cells, LPS produced a >4-fold immediate and sustained increase in CD55 mRNA and protein expression, which was blocked by NF-κB inhibitors. Our results unexpectedly suggest that vitamin D3 signaling may promote an anti-inflammatory response through an NF-κB-dependent increase in CD55 expression. As expected, LPS or 1,25-D3 alone led to sustained increases in CD14 and CD11b expression. In 1,25-D3 pretreated cells, LPS differentially regulated protein expression - CD14 (21-fold increase) and CD11b (a transient 2-fold decrease) - principally at the posttranscriptional level. The coordinated temporal expression of CD55, CD14 and CD11b would contribute to an anti-inflammatory response by providing protection against complement-mediated cell lysis during pathogen recognition and eradication. Overall, the vitamin D3 system may play a role coordinating an anti-inflammatory response pattern of the host complement immune system. This may be particularly important when considering the high rates of preterm births in blacks, a population that exhibits reduced circulating vitamin D3 levels.
Collapse
|
42
|
Regulation of C/EBPβ and resulting functions in cells of the monocytic lineage. Cell Signal 2012; 24:1287-96. [DOI: 10.1016/j.cellsig.2012.02.007] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 02/14/2012] [Indexed: 01/10/2023]
|
43
|
Kim M, Mirandola L, Pandey A, Nguyen DD, Jenkins MR, Turcel M, Cobos E, Chiriva-Internati M. Application of vitamin D and derivatives in hematological malignancies. Cancer Lett 2012; 319:8-22. [DOI: 10.1016/j.canlet.2011.10.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 10/15/2011] [Accepted: 10/17/2011] [Indexed: 11/16/2022]
|
44
|
Cell-Type-Specific Effects of Silibinin on Vitamin D-Induced Differentiation of Acute Myeloid Leukemia Cells Are Associated with Differential Modulation of RXRα Levels. LEUKEMIA RESEARCH AND TREATMENT 2012; 2012:401784. [PMID: 23259067 PMCID: PMC3505927 DOI: 10.1155/2012/401784] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2012] [Accepted: 03/15/2012] [Indexed: 01/22/2023]
Abstract
Plant polyphenols have been shown to enhance the differentiation of acute myeloid leukemia (AML) cells induced by the hormonal form of vitamin D(3) (1α,25-dihydroxyvitamin D(3); 1,25D). However, how these agents modulate 1,25D effects in different subtypes of AML cells remains poorly understood. Here, we show that both carnosic acid (CA) and silibinin (SIL) synergistically enhancd 1,25D-induced differentiation of myeloblastic HL60 cells. However, in promonocytic U937 cells, only CA caused potentiation while SIL attenuated 1,25D effect. The enhanced effect of 1,25D+CA was accompanied by increases in both the vitamin D receptor (VDR) and retinoid X receptor alpha (RXRα) protein levels and vitamin D response element (VDRE) transactivation in both cell lines. Similar increases were observed in HL60 cells treated with 1,25D + SIL. In U937 cells, however, SIL inhibited 1,25D-induced VDRE transactivation concomitant with downregulation of RXRα at both transcriptional and posttranscriptional levels. These inhibitory effects correlated with the inability of SIL, with or without 1,25D, to activate the Nrf2/antioxidant response element signaling pathway in U937 cells. These results suggest that opposite effects of SIL on 1,25D-induced differentiation of HL60 and U937 cells may be determined by cell-type-specific signaling and transcriptional responses to this polyphenol resulting in differential modulation of RXRα expression.
Collapse
|
45
|
Brown G, Hughes PJ. A role for caspase inhibitors in differentiation therapy of myeloid leukaemia. Leuk Res 2012; 36:808-10. [PMID: 22534101 DOI: 10.1016/j.leukres.2012.03.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 03/31/2012] [Accepted: 03/31/2012] [Indexed: 11/29/2022]
|
46
|
Wen CL, Teng CL, Chiang CH, Chang CC, Hwang WL, Kuo CL, Hsu SL. Methanol extract of Antrodia cinnamomea mycelia induces phenotypic and functional differentiation of HL60 into monocyte-like cells via an ERK/CEBP-β signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2012; 19:424-435. [PMID: 22293124 DOI: 10.1016/j.phymed.2011.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 10/05/2011] [Accepted: 11/02/2011] [Indexed: 05/31/2023]
Abstract
Antrodia cinnamomea (named as Niu-chang-chih), a well-known Taiwanese folk medicinal mushroom, has a spectrum of biological activities, especially with anti-tumor property. This study was carried out for the first time to examine the potential role and the underlying mechanisms of A. cinnamomea in the differentiation of human leukemia HL60 cells. We found that the methanol extract of liquid cultured mycelia of A. cinnamomea (MEMAC) inhibited proliferation and induced G1-phase cell cycle arrest in HL60 cells. MEMAC could induce differentiation of HL60 cells into the monocytic lineage, as evaluated by the morphological change, nitroblue tetrazolium reduction assay, non-specific esterase assay, and expression of CD14 and CD11b surface antigens. In addition, MEMAC activated the extracellular signal-regulated kinase (ERK) pathway and increased CCAAT/enhancer-binding protein β (C/EBPβ) expression. Reverse transcriptase polymerase chain reaction analysis showed that MEMAC upregulated the expression of C/EBPβ and CD14 mRNA in HL60 cells. DNA affinity precipitation assay and chromatin immunoprecipitation analyses indicated that MEMAC enhanced the direct binding of C/EBPβ to its response element located at upstream of the CD14 promoter. Furthermore, inhibiting ERK pathway activation with PD98059 markedly blocked MEMAC-induced HL60 monocytic differentiation. Consistently, the MEMAC-mediated upregulation of C/EBPβ and CD14 was also suppressed by PD98059. These findings demonstrate that MEMAC-induced HL60 cell monocytic differentiation is via the activating ERK signaling pathway, and downstream upregulating the transcription factor C/EBPβ and differentiation marker CD14 gene, suggesting that MEMAC might be a potential differentiation-inducing agent for treatment of leukemia.
Collapse
Affiliation(s)
- Chi-Luan Wen
- Taiwan Seed Improvement and Propagation Station, Council of Agriculture, Propagation Technology Section, Taichung, Taiwan
| | | | | | | | | | | | | |
Collapse
|
47
|
Pardanani A, Drake MT, Finke C, Lasho TL, Rozell SA, Jimma T, Tefferi A. Vitamin D insufficiency in myeloproliferative neoplasms and myelodysplastic syndromes: clinical correlates and prognostic studies. Am J Hematol 2011; 86:1013-6. [PMID: 22086864 DOI: 10.1002/ajh.22181] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vitamin D insufficiency is commonly observed in the general population; observational studies have suggested an association with increased risk of cancer development. We examined the clinical and prognostic relevance of low plasma levels of 25-hydroxyvitamin D (25[OH]D) in myeloproliferative neoplasms (MPN) and myelodysplastic syndromes (MDS). A total of 409 patients were studied: 247 (60%) with primary myelofibrosis (PMF), 74 (18%) with de novo MDS, 63 (15%) with polycythemia vera (PV), and 25 (6%) with essential thrombocythemia (ET). Plasma 25(OH)D levels were measured by liquid chromatography-tandem mass spectrometry; a level lower than 25 ng/mL indicated vitamin D insufficiency and a level lower than 10 ng/mL indicated severe deficiency. The proportion of patients with 25(OH)D insufficiency was significantly greater in PMF (48%) and PV (43%) when compared with ET (28%) and MDS (28%) (P = 0.01). Severe 25(OH)D deficiency was significantly more frequent in ET (12%) and PMF (9%), compared with PV (3%) and MDS (1%) (P = 0.05). There were no significant correlations between 25(OH)D insufficiency, or severe deficiency, and a variety of clinical or laboratory variables in PMF, MDS, or PV. Furthermore, Vitamin D insufficiency did not influence either overall or leukemia-free survival in PMF, MDS, or PV (P > 0.05). We conclude that while hypovitaminosis D is relatively common in MPN and MDS, its clinical relevance for prognosis is limited.
Collapse
Affiliation(s)
- Animesh Pardanani
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
MicroRNA-146a and AMD3100, two ways to control CXCR4 expression in acute myeloid leukemias. Blood Cancer J 2011; 1:e26. [PMID: 22829170 PMCID: PMC3255264 DOI: 10.1038/bcj.2011.24] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 05/12/2011] [Indexed: 12/30/2022] Open
Abstract
CXCR4 is a negative prognostic marker in acute myeloid leukemias (AMLs). Therefore, it is necessary to develop novel ways to inhibit CXCR4 expression in leukemia. AMD3100 is an inhibitor of CXCR4 currently used to mobilize cancer cells. CXCR4 is a target of microRNA (miR)-146a that may represent a new tool to inhibit CXCR4 expression. We then investigated CXCR4 regulation by miR-146a in primary AMLs and found an inverse correlation between miR-146a and CXCR4 protein expression levels in all AML subtypes. As the lowest miR-146a expression levels were observed in M5 AML, we analyzed the control of CXCR4 expression by miR-146a in normal and leukemic monocytic cells and showed that the regulatory miR-146a/CXCR4 pathway operates during monocytopoiesis, but is deregulated in AMLs. AMD3100 treatment and miR-146a overexpression were used to inhibit CXCR4 in leukemic cells. AMD3100 treatment induces the decrease of CXCR4 protein expression, associated with miR-146a increase, and increases sensitivity of leukemic blast cells to cytotoxic drugs, this effect being further enhanced by miR-146a overexpression. Altogether our data indicate that miR-146a and AMD3100, acting through different mechanism, downmodulate CXCR4 protein levels, impair leukemic cell proliferation and then may be used in combination with anti-leukemia drugs, for development of new therapeutic strategies.
Collapse
|
49
|
Gocek E, Marcinkowska E. Differentiation therapy of acute myeloid leukemia. Cancers (Basel) 2011; 3:2402-20. [PMID: 24212816 PMCID: PMC3757424 DOI: 10.3390/cancers3022402] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2011] [Revised: 04/29/2011] [Accepted: 05/05/2011] [Indexed: 12/31/2022] Open
Abstract
Acute Myeloid Leukemia (AML) is a predominant acute leukemia among adults, characterized by accumulation of malignantly transformed immature myeloid precursors. A very attractive way to treat myeloid leukemia, which is now called 'differentiation therapy', was proposed as in vitro studies have shown that a variety of agents stimulate differentiation of the cell lines isolated from leukemic patients. One of the differentiation-inducing agents, all-trans retinoic acid (ATRA), which can induce granulocytic differentiation in myeloid leukemic cell lines, has been introduced into clinics to treat patients with acute promyelocytic leukemia (APL) in which a PML-RARA fusion protein is generated by a t(15;17)(q22;q12) chromosomal translocation. Because differentiation therapy using ATRA has significantly improved prognosis for patients with APL, many efforts have been made to find alternative differentiating agents. Since 1,25-dihydroxyvitamin D3 (1,25D) is capable of inducing in vitro monocyte/macrophage differentiation of myeloid leukemic cells, clinical trials have been performed to estimate its potential to treat patients with AML or myelodysplastic syndrome (MDS). Unfortunately therapeutic concentrations of 1,25D can induce potentially fatal systemic hypercalcemia, thus limiting clinical utility of that compound. Attempts to overcome this problem have focused on the synthesis of 1,25D analogs (VDAs) which retain differentiation inducing potential, but lack its hypercalcemic effects. This review aims to discuss current problems and potential solutions in differentiation therapy of AML.
Collapse
Affiliation(s)
- Elzbieta Gocek
- Department of Biotechnology, University of Wroclaw, ul Tamka 2, Wroclaw 50-137, Poland; E-Mail: (E.G.)
| | - Ewa Marcinkowska
- Department of Biotechnology, University of Wroclaw, ul Tamka 2, Wroclaw 50-137, Poland; E-Mail: (E.G.)
| |
Collapse
|
50
|
Musgrove EA, Sutherland RL. RB in breast cancer: differential effects in estrogen receptor-positive and estrogen receptor-negative disease. Cell Cycle 2011; 9:4607. [PMID: 21260944 DOI: 10.4161/cc.9.23.13889] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|