1
|
Stajer M, Horacek JM, Kupsa T, Zak P. The role of chemokines and interleukins in acute lymphoblastic leukemia: a systematic review. J Appl Biomed 2024; 22:165-184. [PMID: 40033805 DOI: 10.32725/jab.2024.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/15/2024] [Indexed: 03/05/2025] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood hematological malignancy, but it also affects adult patients with worse prognosis and outcomes. Leukemic cells benefit from protective mechanisms, which are mediated by intercellular signaling molecules - cytokines. Through these signals, cytokines modulate the biology of leukemic cells and their surroundings, enhancing the proliferation, survival, and chemoresistance of the disease. This ultimately leads to disease progression, refractoriness, and relapse, decreasing the chances of curability and overall survival of the patients. Targeting and modulating these pathological processes without affecting the healthy physiology is desirable, offering more possibilities for the treatment of ALL patients, which still remains unsatisfactory in certain cases. In this review, we comprehensively analyze the existing literature and ongoing trials regarding the role of chemokines and interleukins in the biology of ALL. Focusing on the functional pathways, genetic background, and critical checkpoints, we constructed a summary of molecules that are promising for prognostic stratification and mainly therapeutic use. Targeted therapy, including chemokine and interleukin pathways, is a new and promising approach to the treatment of cancer. With the expansion of our knowledge, we are able to uncover a spectrum of new potential checkpoints in order to modulate the disease biology. Several cytokine-related targets are advancing toward clinical application, offering the hope of higher disease response rates to treatment.
Collapse
Affiliation(s)
- Martin Stajer
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Jan M Horacek
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Tomas Kupsa
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Pavel Zak
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| |
Collapse
|
2
|
Ma Z, Zhou F, Jin H, Wu X. Crosstalk between CXCL12/CXCR4/ACKR3 and the STAT3 Pathway. Cells 2024; 13:1027. [PMID: 38920657 PMCID: PMC11201928 DOI: 10.3390/cells13121027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
The reciprocal modulation between the CXCL12/CXCR4/ACKR3 axis and the STAT3 signaling pathway plays a crucial role in the progression of various diseases and neoplasms. Activation of the CXCL12/CXCR4/ACKR3 axis triggers the STAT3 pathway through multiple mechanisms, while the STAT3 pathway also regulates the expression of CXCL12. This review offers a thorough and systematic analysis of the reciprocal regulatory mechanisms between the CXCL12/CXCR4/ACKR3 signaling axis and the STAT3 signaling pathway in the context of diseases, particularly tumors. It explores the potential clinical applications in tumor treatment, highlighting possible therapeutic targets and novel strategies for targeted tumor therapy.
Collapse
Affiliation(s)
| | | | | | - Xiaoming Wu
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming 650500, China; (Z.M.); (F.Z.); (H.J.)
| |
Collapse
|
3
|
Naser IH, Hamza AA, Alhili A, Faisal AN, Ali MS, Kadhim NA, Suliman M, Alshahrani MY, Alawadi A. Atypical chemokine receptor 4 (ACKR4/CCX-CKR): A comprehensive exploration across physiological and pathological landscapes in contemporary research. Cell Biochem Funct 2024; 42:e4009. [PMID: 38597217 DOI: 10.1002/cbf.4009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/02/2024] [Accepted: 03/31/2024] [Indexed: 04/11/2024]
Abstract
Atypical chemokine receptor 4 (ACKR4), also known as CCX-CKR, is a member of the chemokine receptor family that lacks typical G protein signaling activity. Instead, ACKR4 functions as a scavenger receptor that can bind and internalize a wide range of chemokines, influencing their availability and activity in the body. ACKR4 is involved in various physiological processes, such as immune cell trafficking and the development of thymus, spleen, and lymph nodes. Moreover, ACKR4 has been implicated in several pathological conditions, including cancer, heart and lung diseases. In cancer, ACKR4 plays a complex role, acting as a tumor suppressor or promoter depending on the type of cancer and the stage of the disease. For instance, ACKR4 may inhibit the growth and metastasis of breast cancer, but it may also promote the progression of hepatocellular carcinoma and gastric cancer. In inflammatory situations, ACKR4 has been found to modulate the recruitment and activation of immune cells, contributing to the pathogenesis of diseases such as myocardial infraction and pulmonary sarcoidosis. The study of ACKR4 is still ongoing, and further research is needed to fully understand its role in different physiological and pathological contexts. Nonetheless, ACKR4 represents a promising target for the development of novel therapeutic strategies for various diseases.
Collapse
Affiliation(s)
- Israa Habeeb Naser
- Medical Laboratories Techniques Department, AL-Mustaqbal University College, Hillah, Babil, Iraq
| | - Asia Ali Hamza
- Department of Pharmaceutics, Faculty of pharmacy, University of Al-Ameed, Karbala, Iraq
| | - Ahmed Alhili
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | | | | | | - Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Ahmed Alawadi
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
4
|
Saluja S, Bansal I, Bhardwaj R, Beg MS, Palanichamy JK. Inflammation as a driver of hematological malignancies. Front Oncol 2024; 14:1347402. [PMID: 38571491 PMCID: PMC10987768 DOI: 10.3389/fonc.2024.1347402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/05/2024] [Indexed: 04/05/2024] Open
Abstract
Hematopoiesis is a tightly regulated process that produces all adult blood cells and immune cells from multipotent hematopoietic stem cells (HSCs). HSCs usually remain quiescent, and in the presence of external stimuli like infection or inflammation, they undergo division and differentiation as a compensatory mechanism. Normal hematopoiesis is impacted by systemic inflammation, which causes HSCs to transition from quiescence to emergency myelopoiesis. At the molecular level, inflammatory cytokine signaling molecules such as tumor necrosis factor (TNF), interferons, interleukins, and toll-like receptors can all cause HSCs to multiply directly. These cytokines actively encourage HSC activation, proliferation, and differentiation during inflammation, which results in the generation and activation of immune cells required to combat acute injury. The bone marrow niche provides numerous soluble and stromal cell signals, which are essential for maintaining normal homeostasis and output of the bone marrow cells. Inflammatory signals also impact this bone marrow microenvironment called the HSC niche to regulate the inflammatory-induced hematopoiesis. Continuous pro-inflammatory cytokine and chemokine activation can have detrimental effects on the hematopoietic system, which can lead to cancer development, HSC depletion, and bone marrow failure. Reactive oxygen species (ROS), which damage DNA and ultimately lead to the transformation of HSCs into cancerous cells, are produced due to chronic inflammation. The biological elements of the HSC niche produce pro-inflammatory cytokines that cause clonal growth and the development of leukemic stem cells (LSCs) in hematological malignancies. The processes underlying how inflammation affects hematological malignancies are still not fully understood. In this review, we emphasize the effects of inflammation on normal hematopoiesis, the part it plays in the development and progression of hematological malignancies, and potential therapeutic applications for targeting these pathways for therapy in hematological malignancies.
Collapse
|
5
|
Rafique A, Hichiwa G, Jatnika MF, Ito Y. A Novel Strategy for Screening Tumor-Specific Variable Domain of Heavy-Chain Antibodies. Int J Mol Sci 2023; 24:10804. [PMID: 37445977 DOI: 10.3390/ijms241310804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
The properties of the variable domain of heavy-chain (VHH) antibodies are particularly relevant in cancer therapy. To isolate tumor cell-specific VHH antibodies, VHH phage libraries were constructed from multiple tumor cells. After enriching the libraries against particular tumor cell lines, a next-generation sequencer was used to screen the pooled phages of each library for potential antibody candidates. Based on high amplification folds, 50 sequences from each library were used to construct phylogenetic trees. Several clusters with identical CDR3 were observed. Groups X, Y, and Z were assigned as common sequences among the different trees. These identical groups over the trees were considered to be cross-reactive antibodies. To obtain monoclonal antibodies, we assembled 200 sequences (top 50 sequences from each library) and rebuilt a combined molecular phylogenetic tree. Groups were categorized as A-G. For each group, we constructed a phagemid and determined its binding specificity with tumor cells. The phage-binding results were consistent with the phylogenetic tree-generated groups, which indicated particular tumor-specific clusters; identical groups showed cross-reactivity. The strategy used in the current study is effective for screening and isolating monoclonal antibodies. Specific antibodies can be identified, even when the target markers of cancer cells are unknown.
Collapse
Affiliation(s)
- Abdur Rafique
- Graduate School of Science and Engineering, University of Kagoshima, Kagoshima 890-0065, Japan
| | - Genki Hichiwa
- Graduate School of Medical Sciences, Tottori University, Tottori 680-8550, Japan
| | - Muhammad Feisal Jatnika
- Graduate School of Science and Engineering, University of Kagoshima, Kagoshima 890-0065, Japan
| | - Yuji Ito
- Graduate School of Science and Engineering, University of Kagoshima, Kagoshima 890-0065, Japan
| |
Collapse
|
6
|
Yang Y, Li J, Lei W, Wang H, Ni Y, Liu Y, Yan H, Tian Y, Wang Z, Yang Z, Yang S, Yang Y, Wang Q. CXCL12-CXCR4/CXCR7 Axis in Cancer: from Mechanisms to Clinical Applications. Int J Biol Sci 2023; 19:3341-3359. [PMID: 37497001 PMCID: PMC10367567 DOI: 10.7150/ijbs.82317] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 05/16/2023] [Indexed: 07/28/2023] Open
Abstract
Cancer is a multi-step disease caused by the accumulation of genetic mutations and/or epigenetic changes, and is the biggest challenge around the world. Cytokines, including chemokines, exhibit expression changes and disorders in all human cancers. These cytokine abnormalities can disrupt homeostasis and immune function, and make outstanding contributions to various stages of cancer development such as invasion, metastasis, and angiogenesis. Chemokines are a superfamily of small molecule chemoattractive cytokines that mediate a variety of cellular functions. Importantly, the interactions of chemokine members CXCL12 and its receptors CXCR4 and CXCR7 have a broad impact on tumor cell proliferation, survival, angiogenesis, metastasis, and tumor microenvironment, and thus participate in the onset and development of many cancers including leukemia, breast cancer, lung cancer, prostate cancer and multiple myeloma. Therefore, this review aims to summarize the latest research progress and future challenges regarding the role of CXCL12-CXCR4/CXCR7 signaling axis in cancer, and highlights the potential of CXCL12-CXCR4/CXCR7 as a biomarker or therapeutic target for cancer, providing essential strategies for the development of novel targeted cancer therapies.
Collapse
Affiliation(s)
- Yaru Yang
- Department of Orthopedics, Shenmu Hospital, Faculty of Life Sciences and Medicine, Northwest University, Shenmu, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Jiayan Li
- Department of Orthopedics, Shenmu Hospital, Faculty of Life Sciences and Medicine, Northwest University, Shenmu, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Wangrui Lei
- Department of Orthopedics, Shenmu Hospital, Faculty of Life Sciences and Medicine, Northwest University, Shenmu, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Haiying Wang
- Department of Orthopedics, Shenmu Hospital, Faculty of Life Sciences and Medicine, Northwest University, Shenmu, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yunfeng Ni
- Department of Thoracic Surgery, Tangdu Hospital, The Airforce Medical University, Xi'an, China
| | - Yanqing Liu
- Department of Orthopedics, Shenmu Hospital, Faculty of Life Sciences and Medicine, Northwest University, Shenmu, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Huanle Yan
- Department of Orthopedics, Shenmu Hospital, Faculty of Life Sciences and Medicine, Northwest University, Shenmu, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yifan Tian
- Department of Orthopedics, Shenmu Hospital, Faculty of Life Sciences and Medicine, Northwest University, Shenmu, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Zhi Yang
- Department of Thoracic Surgery, Tangdu Hospital, The Airforce Medical University, Xi'an, China
| | - Shulin Yang
- Department of Orthopedics, Shenmu Hospital, Faculty of Life Sciences and Medicine, Northwest University, Shenmu, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yang Yang
- Department of Orthopedics, Shenmu Hospital, Faculty of Life Sciences and Medicine, Northwest University, Shenmu, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Qiang Wang
- Department of Orthopedics, Shenmu Hospital, Faculty of Life Sciences and Medicine, Northwest University, Shenmu, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| |
Collapse
|
7
|
Bueno MLP, Saad STO, Roversi FM. The antitumor effects of WNT5A against hematological malignancies. J Cell Commun Signal 2023:10.1007/s12079-023-00773-8. [PMID: 37310653 DOI: 10.1007/s12079-023-00773-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/30/2023] [Indexed: 06/14/2023] Open
Abstract
The bone marrow (BM) microenvironment (niche) is abnormally altered in acute myeloid leukemia (AML), leading to deficient secretion of proteins, soluble factors, and cytokines by mesenchymal stromal cells (MSC) that modifies the crosstalk between MSC and hematopoietic cells. We focused on a WNT gene/protein family member, WNT5A, which is downregulated in leukemia and correlated with disease progression and poor prognosis. We demonstrated that WNT5A protein upregulated the WNT non-canonical pathway only in leukemic cells, without modulating normal cell behavior. We also introduced a novel WNT5A-mimicking compound, Foxy-5. Our results showed reduction of crucial biological functions that are upregulated in leukemia cells, including ROS generation, cell proliferation, and autophagy, as well as G0/G1 cell cycle arrest. Additionally, Foxy-5 induced early-stage macrophage cell differentiation, a crucial process during leukemia development. At a molecular level, Foxy-5 led to the downregulation of two overexpressed leukemia pathways, PI3K and MAPK, which resulted in a disarrangement of actin polymerization with consequent impairment of CXCL12-induced chemotaxis. Notably, in a novel tri-dimensional bone marrow-mimicking model, Foxy-5 led to reduced leukemia cell growth and similar results were observed in a xenograft in vivo model. Overall, our findings highlight the pivotal role of WNT5A in leukemia and demonstrate that Foxy-5 acts as a specific antineoplastic agent in leukemia, counterbalancing several leukemic oncogenic processes related to the crosstalk in the bone marrow niche, and represents a promising therapeutic option for AML. WNT5A, a WNT gene/protein family member, is naturally secreted by mesenchymal stromal cells and contributes to the maintenance of the bone marrow microenvironment. WNT5A downregulation is correlated with disease progression and poor prognosis. The treatment with Foxy-5, a WNT5A mimetizing compound, counterbalanced several leukemogenic processes that are upregulated in leukemia cells, including ROS generation, cell proliferation, and autophagy and disruption of PI3K and MAPK signaling pathways.
Collapse
Affiliation(s)
- Maura Lima Pereira Bueno
- Hematology and Transfusion Medicine Center - University of Campinas/Hemocentro-UNICAMP, Rua Carlos Chagas, 480 - Cidade Universitária Zeferino Vaz - Barão Geraldo, Campinas, São Paulo, 13083-878, Brazil
| | - Sara Teresinha Olalla Saad
- Hematology and Transfusion Medicine Center - University of Campinas/Hemocentro-UNICAMP, Rua Carlos Chagas, 480 - Cidade Universitária Zeferino Vaz - Barão Geraldo, Campinas, São Paulo, 13083-878, Brazil
| | - Fernanda Marconi Roversi
- Hematology and Transfusion Medicine Center - University of Campinas/Hemocentro-UNICAMP, Rua Carlos Chagas, 480 - Cidade Universitária Zeferino Vaz - Barão Geraldo, Campinas, São Paulo, 13083-878, Brazil.
- Department of Surgery Division of Transplantation, Emory University, Atlanta, GA, USA.
| |
Collapse
|
8
|
Antonello P, Pizzagalli DU, Foglierini M, Melgrati S, Radice E, Thelen S, Thelen M. ACKR3 promotes CXCL12/CXCR4-mediated cell-to-cell-induced lymphoma migration through LTB4 production. Front Immunol 2023; 13:1067885. [PMID: 36713377 PMCID: PMC9878562 DOI: 10.3389/fimmu.2022.1067885] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/22/2022] [Indexed: 01/14/2023] Open
Abstract
Chemotaxis is an essential physiological process, often harnessed by tumors for metastasis. CXCR4, its ligand CXCL12 and the atypical receptor ACKR3 are overexpressed in many human cancers. Interfering with this axis by ACKR3 deletion impairs lymphoma cell migration towards CXCL12. Here, we propose a model of how ACKR3 controls the migration of the diffused large B-cell lymphoma VAL cells in vitro and in vivo in response to CXCL12. VAL cells expressing full-length ACKR3, but not a truncated version missing the C-terminus, can support the migration of VAL cells lacking ACKR3 (VAL-ko) when allowed to migrate together. This migration of VAL-ko cells is pertussis toxin-sensitive suggesting the involvement of a Gi-protein coupled receptor. RNAseq analysis indicate the expression of chemotaxis-mediating LTB4 receptors in VAL cells. We found that LTB4 acts synergistically with CXCL12 in stimulating the migration of VAL cells. Pharmacologic or genetic inhibition of BLT1R markedly reduces chemotaxis towards CXCL12 suggesting that LTB4 enhances in a contact-independent manner the migration of lymphoma cells. The results unveil a novel mechanism of cell-to-cell-induced migration of lymphoma.
Collapse
Affiliation(s)
- Paola Antonello
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Graduate School of Cellular and Molecular Sciences, University of Bern, Bern, Switzerland
| | - Diego U. Pizzagalli
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Università della Svizzera italiana, Euler Institute, Lugano-Viganello, Switzerland
| | - Mathilde Foglierini
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Serena Melgrati
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Graduate School of Cellular and Molecular Sciences, University of Bern, Bern, Switzerland
| | - Egle Radice
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Sylvia Thelen
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Marcus Thelen
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| |
Collapse
|
9
|
Perbellini O, Cavallini C, Chignola R, Galasso M, Scupoli MT. Phospho-Specific Flow Cytometry Reveals Signaling Heterogeneity in T-Cell Acute Lymphoblastic Leukemia Cell Lines. Cells 2022; 11:cells11132072. [PMID: 35805156 PMCID: PMC9266179 DOI: 10.3390/cells11132072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/15/2022] [Accepted: 06/27/2022] [Indexed: 12/10/2022] Open
Abstract
Several signaling pathways are aberrantly activated in T-ALL due to genetic alterations of their components and in response to external microenvironmental cues. To functionally characterize elements of the signaling network in T-ALL, here we analyzed ten signaling proteins that are frequently altered in T-ALL -namely Akt, Erk1/2, JNK, Lck, NF-κB p65, p38, STAT3, STAT5, ZAP70, Rb- in Jurkat, CEM and MOLT4 cell lines, using phospho-specific flow cytometry. Phosphorylation statuses of signaling proteins were measured in the basal condition or under modulation with H2O2, PMA, CXCL12 or IL7. Signaling profiles are characterized by a high variability across the analyzed T-ALL cell lines. Hierarchical clustering analysis documents that higher intrinsic phosphorylation of Erk1/2, Lck, ZAP70, and Akt, together with ZAP70 phosphorylation induced by H2O2, identifies Jurkat cells. In contrast, CEM are characterized by higher intrinsic phosphorylation of JNK and Rb and higher responsiveness of Akt to external stimuli. MOLT4 cells are characterized by higher basal STAT3 phosphorylation. These data document that phospho-specific flow cytometry reveals a high variability in intrinsic as well as modulated signaling networks across different T-ALL cell lines. Characterizing signaling network profiles across individual leukemia could provide the basis to identify molecular targets for personalized T-ALL therapy.
Collapse
Affiliation(s)
- Omar Perbellini
- Department of Cell Therapy and Hematology, San Bortolo Hospital, Viale Ferdinando Rodolfi, 37, 36100 Vicenza, Italy;
| | - Chiara Cavallini
- Research Center LURM, Interdepartmental Laboratory of Medical Research, University of Verona, Piazzale L.A. Scuro, 10, 37134 Verona, Italy;
| | - Roberto Chignola
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134 Verona, Italy;
| | - Marilisa Galasso
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Piazzale L.A. Scuro, 10, 37134 Verona, Italy;
| | - Maria T. Scupoli
- Research Center LURM, Interdepartmental Laboratory of Medical Research, University of Verona, Piazzale L.A. Scuro, 10, 37134 Verona, Italy;
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Piazzale L.A. Scuro, 10, 37134 Verona, Italy;
- Correspondence: ; Tel.: +39-045-8128-425
| |
Collapse
|
10
|
Mehrpouri M. The contributory roles of the CXCL12/CXCR4/CXCR7 axis in normal and malignant hematopoiesis: A possible therapeutic target in hematologic malignancies. Eur J Pharmacol 2022; 920:174831. [PMID: 35183534 DOI: 10.1016/j.ejphar.2022.174831] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/03/2022]
|
11
|
Madrazo E, González-Novo R, Ortiz-Placín C, García de Lacoba M, González-Murillo Á, Ramírez M, Redondo-Muñoz J. Fast H3K9 methylation promoted by CXCL12 contributes to nuclear changes and invasiveness of T-acute lymphoblastic leukemia cells. Oncogene 2022; 41:1324-1336. [PMID: 34999734 DOI: 10.1038/s41388-021-02168-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 12/09/2021] [Accepted: 12/22/2021] [Indexed: 02/06/2023]
Abstract
T-acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy that comprises the accumulation of malignant T-cells. Despite current therapies, failure to conventional treatments and relapse are frequent in children with T-ALL. It is known that the chemokine CXCL12 modulates leukemia survival and dissemination; however, our understanding of molecular mechanisms used by T-ALL cells to infiltrate and respond to leukemia cells-microenvironment interactions is still vague. In the present study, we showed that CXCL12 promoted H3K9 methylation in cell lines and primary T-ALL cells within minutes. We thus identified that CXCL12-mediated H3K9 methylation affected the global chromatin configuration and the nuclear mechanics of T-ALL cells. Importantly, we characterized changes in the genomic profile of T-ALL cells associated with rapid CXCL12 stimulation. We showed that blocking CXCR4 and protein kinase C (PKC) impaired the H3K9 methylation induced by CXCL12 in T-ALL cells. Finally, blocking H3K9 methyltransferases reduced the efficiency of T-ALL cells to deform their nuclei, migrate across confined spaces, and home to spleen and bone marrow in vivo models. Together, our data show novel functions for CXL12 as a master regulator of nuclear deformability and epigenetic changes in T-ALL cells, and its potential as a promising pharmacological target against T-ALL dissemination.
Collapse
Affiliation(s)
- Elena Madrazo
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Raquel González-Novo
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Cándido Ortiz-Placín
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Mario García de Lacoba
- Bioinformatics and Biostatistics Unit, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - África González-Murillo
- Department of Paediatric Haematology & Oncology, Hospital Universitario Niño Jesús, Madrid, Spain
- Health Research Institute La Princesa, Madrid, Spain
| | - Manuel Ramírez
- Department of Paediatric Haematology & Oncology, Hospital Universitario Niño Jesús, Madrid, Spain
- Health Research Institute La Princesa, Madrid, Spain
| | - Javier Redondo-Muñoz
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain.
| |
Collapse
|
12
|
Sottoriva K, Pajcini KV. Notch Signaling in the Bone Marrow Lymphopoietic Niche. Front Immunol 2021; 12:723055. [PMID: 34394130 PMCID: PMC8355626 DOI: 10.3389/fimmu.2021.723055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Lifelong mammalian hematopoiesis requires continuous generation of mature blood cells that originate from Hematopoietic Stem and Progenitor Cells (HSPCs) situated in the post-natal Bone Marrow (BM). The BM microenvironment is inherently complex and extensive studies have been devoted to identifying the niche that maintains HSPC homeostasis and supports hematopoietic potential. The Notch signaling pathway is required for the emergence of the definitive Hematopoietic Stem Cell (HSC) during embryonic development, but its role in BM HSC homeostasis is convoluted. Recent work has begun to explore novel roles for the Notch signaling pathway in downstream progenitor populations. In this review, we will focus an important role for Notch signaling in the establishment of a T cell primed sub-population of Common Lymphoid Progenitors (CLPs). Given that its activation mechanism relies primarily on cell-to-cell contact, Notch signaling is an ideal means to investigate and define a novel BM lymphopoietic niche. We will discuss how new genetic model systems indicate a pre-thymic, BM-specific role for Notch activation in early T cell development and what this means to the paradigm of lymphoid lineage commitment. Lastly, we will examine how leukemic T-cell acute lymphoblastic leukemia (T-ALL) blasts take advantage of Notch and downstream lymphoid signals in the pathological BM niche.
Collapse
Affiliation(s)
- Kilian Sottoriva
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, United States
| | - Kostandin V Pajcini
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, United States
| |
Collapse
|
13
|
Roversi FM, Bueno MLP, Pericole FV, Saad STO. Hematopoietic Cell Kinase (HCK) Is a Player of the Crosstalk Between Hematopoietic Cells and Bone Marrow Niche Through CXCL12/CXCR4 Axis. Front Cell Dev Biol 2021; 9:634044. [PMID: 33842460 PMCID: PMC8027121 DOI: 10.3389/fcell.2021.634044] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 02/26/2021] [Indexed: 12/12/2022] Open
Abstract
The crosstalk between hematopoietic stem/progenitor cells (HSC), both normal and leukemic, and their neighboring bone marrow (BM) microenvironment (niche) creates a reciprocal dependency, a master regulator of biological process, and chemotherapy resistance. In acute myeloid leukemia (AML), leukemic stem/progenitor cells (LSC) anchored in the protective BM microenvironment, reprogram and transform this niche into a leukemia-supporting and chemoprotective environment. One most important player involved in this crosstalk are CXCL12, produced by the BM mesenchymal stromal cells, and its receptor CXCR4, present onto HSC. The downstream molecular mechanisms involved in CXCL12/CXCR4 axis have many targets, including the Src family members of non-receptor tyrosine kinase (SFK). We herein study the role of one SFK member, the Hematopoietic Cell Kinase (HCK), in CXCL12/CXCR4 pathway and its contribution to the AML pathogenesis. We verified that the inhibition of HCK severely impaired CXCL12-induced migration of leukemic cell lines and CD34 positive cells from AML patients bone marrow, through a disruption of the activation of CXCL12/CXCR4/PI3K/AKT and CXCL12/CXCR4/MAPK/ERK signaling, and by a decreased cytoskeleton dynamic through a lower rate of actin polymerization. We provide new insights into the key role of HCK in conferring a migratory advantage to leukemic cells thought CXCL12/CXCR4 axis. HCK represents an important protein of the main pathway involved in the crosstalk between HSC, and their surrounding milieu. Thus, HCK inhibition could represent a novel approach for the treatment of the acute myeloid leukemia.
Collapse
Affiliation(s)
- Fernanda Marconi Roversi
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil
| | - Maura Lima Pereira Bueno
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil
| | - Fernando Viera Pericole
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil
| | - Sara Teresinha Olalla Saad
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil
| |
Collapse
|
14
|
Hong Z, Wei Z, Xie T, Fu L, Sun J, Zhou F, Jamal M, Zhang Q, Shao L. Targeting chemokines for acute lymphoblastic leukemia therapy. J Hematol Oncol 2021; 14:48. [PMID: 33743810 PMCID: PMC7981899 DOI: 10.1186/s13045-021-01060-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a hematological malignancy characterized by the malignant clonal expansion of lymphoid hematopoietic precursors. It is regulated by various signaling molecules such as cytokines and adhesion molecules in its microenvironment. Chemokines are chemotactic cytokines that regulate migration, positioning and interactions of cells. Many chemokine axes such as CXCL12/CXCR4 and CCL25/CCR9 have been proved to play important roles in leukemia microenvironment and further affect ALL outcomes. In this review, we summarize the chemokines that are involved in ALL progression and elaborate on their roles and mechanisms in leukemia cell proliferation, infiltration, drug resistance and disease relapse. We also discuss the potential of targeting chemokine axes for ALL treatments, since many related inhibitors have shown promising efficacy in preclinical trials, and some of them have entered clinical trials.
Collapse
Affiliation(s)
- Zixi Hong
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zimeng Wei
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Tian Xie
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Lin Fu
- The First Clinical School of Wuhan University, Wuhan, China
| | - Jiaxing Sun
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Muhammad Jamal
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Qiuping Zhang
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, China.
| | - Liang Shao
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
15
|
Nguyen HT, Reyes-Alcaraz A, Yong HJ, Nguyen LP, Park HK, Inoue A, Lee CS, Seong JY, Hwang JI. CXCR7: a β-arrestin-biased receptor that potentiates cell migration and recruits β-arrestin2 exclusively through Gβγ subunits and GRK2. Cell Biosci 2020; 10:134. [PMID: 33292475 PMCID: PMC7686738 DOI: 10.1186/s13578-020-00497-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Some chemokine receptors referred to as atypical chemokine receptors (ACKRs) are thought to non-signaling decoys because of their inability to activate typical G-protein signaling pathways. CXCR7, also known as ACKR3, binds to only two chemokines, SDF-1α and I-TAC, and recruits β-arrestins. SDF-1α also binds to its own conventional receptor, CXCR4, involving in homeostatic modulation such as development and immune surveillance as well as pathological conditions such as inflammation, ischemia, and cancers. Recently, CXCR7 is suggested as a key therapeutic target together with CXCR4 in such conditions. However, the molecular mechanisms underlying cellular responses and functional relation with CXCR7 and CXCR4 have not been elucidated, despite massive studies. Therefore, we aimed to reveal the molecular networks of CXCR7 and CXCR4 and compare their effects on cell migration. METHODS Base on structural complementation assay using NanoBiT technology, we characterized the distinct mechanisms underlying β-arrestin2 recruitment by both CXCR4 and CXCR7. Crosslinking and immunoprecipitation were conducted to analyze complex formation of the receptors. Gene deletion using CRISPR and reconstitution of the receptors were applied to analysis of ligand-dependent ERK phosphorylation and cell migration. All experiments were performed in triplicate and repeated more than three times. Unpaired Student's t-tests or ANOVA using PRISM5 software were employed for statistical analyses. RESULTS Ligand binding to CXCR7 does not result in activation of typical signaling pathways via Gα subunits but activation of GRK2 via βγ subunits and receptor phosphorylation with subsequent β-arrestin2 recruitment. In contrast, CXCR4 induced Gαi activation and recruited β-arrestin2 through C-terminal phosphorylation by both GRK2 and GRK5. SDF-1α-stimulated ERK phosphorylation was facilitated by CXCR4, but not CXCR7. Heterodimerization of CXCR4 and CXCR7 was not confirmed in this study, while homodimerization of them was verified by crosslinking experiment and NanoBiT assay. Regarding chemotaxis, SDF-1α-stimulated cell migration was mediated by both CXCR4 and CXCR7. CONCLUSION This study demonstrates that SDF-1α-stimulated CXCR7 mediates β-arrestin2 recruitment via different molecular networking from that of CXCR4. CXCR7 may be neither a simple scavenger nor auxiliary receptor but plays an essential role in cell migration through cooperation with CXCR4.
Collapse
Affiliation(s)
- Huong Thi Nguyen
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | | | - Hyo Jeong Yong
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Lan Phuong Nguyen
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Hee-Kyung Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Cheol Soon Lee
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jae Young Seong
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jong-Ik Hwang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Koch C, Engele J. Functions of the CXCL12 Receptor ACKR3/CXCR7-What Has Been Perceived and What Has Been Overlooked. Mol Pharmacol 2020; 98:577-585. [PMID: 32883765 DOI: 10.1124/molpharm.120.000056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/31/2020] [Indexed: 12/18/2022] Open
Abstract
The CXCL12 system is central to the development of many organs and is further crucially engaged in pathophysiological processes underlying cancer, inflammation, and cardiovascular disorders. This disease-associated role presently focuses major interest on the two CXCL12 receptors, CXCR4 and atypical chemokine receptor 3 (ACKR3)/CXCR7, as promising therapeutic targets. Major obstacles in these ongoing efforts are confusing reports on the differential use of either ACKR3/CXCR7 and/or CXCR4 across various cells as well as on the specific function(s) of ACKR3/CXCR7. Although basically no doubts remain that CXCR4 represents a classic chemokine receptor, functions assigned to ACKR3/CXCR7 range from those of a strictly silent scavenger receptor eventually modulating CXCR4 signaling to an active and independent signaling receptor. In this review, we depict a thorough analysis of our present knowledge on different modes of organization and functions of the cellular CXCL12 system. We further highlight the potential role of ACKR3/CXCR7 as a "crosslinker" of different receptor systems. Finally, we discuss mechanisms with the potency to impinge on the cellular organization of the CXCL12 system and hence might represent additional future therapeutic targets. SIGNIFICANCE STATEMENT: Delineating the recognized functions of atypical chemokine receptor 3 and CXCR4 in CXCL12 signaling is central to the more detailed understanding of the role of the CXCL12 system in health and disease and will help to guide future research efforts.
Collapse
Affiliation(s)
- Christian Koch
- Institute of Anatomy, University of Leipzig, Medical Faculty, Leipzig, Germany
| | - Jürgen Engele
- Institute of Anatomy, University of Leipzig, Medical Faculty, Leipzig, Germany
| |
Collapse
|
17
|
Sklarz LM, Gladbach YS, Ernst M, Hamed M, Roolf C, Sender S, Beck J, Schütz E, Fischer S, Struckmann S, Junghanss C, Fuellen G, Murua Escobar H. Combination of the PI3K inhibitor Idelalisib with the conventional cytostatics cytarabine and dexamethasone leads to changes in pathway activation that induce anti-proliferative effects in B lymphoblastic leukaemia cell lines. Cancer Cell Int 2020; 20:390. [PMID: 32817744 PMCID: PMC7425054 DOI: 10.1186/s12935-020-01431-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 07/16/2020] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND The introduction of combined conventional cytostatics and pathway-specific inhibitors has opened new treatment options for several cancer types including hematologic neoplasia such as leukaemias. As the detailed understanding of the combination-induced molecular effects is often lacking, the identification of combination-induced molecular mechanisms bears significant value for the further development of interventional approaches. METHODS Combined application of conventional cytostatic agents (cytarabine and dexamethasone) with the PI3K-inhibitor Idelalisib was analysed on cell-biologic parameters in two acute pro-B lymphoblastic leukaemia (B-ALL) cell lines. In particular, for comparative characterisation of the molecular signatures induced by the combined and mono application, whole transcriptome sequencing was performed. Emphasis was placed on pathways and genes exclusively regulated by drug combinations. RESULTS Idelalisib + cytostatics combinations changed pathway activation for, e.g., "Retinoblastoma in cancer", "TGF-b signalling", "Cell cycle" and "DNA-damage response" to a greater extent than the two cytostatics alone. Analyses of the top-20 regulated genes revealed that both combinations induce characteristic gene expression changes. CONCLUSION A specific set of genes was exclusively deregulated by the drug combinations, matching the combination-specific anti-proliferative cell-biologic effects. The addition of Idelalisib suggests minor synergistic effects which are rather to be classified as additive.
Collapse
Affiliation(s)
- L.-M. Sklarz
- Department of Medicine, Clinic III - Hematology/Oncology/Palliative Care, Rostock University Medical Center, Rostock, Germany
| | - Y. S. Gladbach
- Institute for Biostatistics and Informatics in Medicine and Ageing Research (IBIMA), Rostock University Medical Center, Rostock, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - M. Ernst
- Institute for Biostatistics and Informatics in Medicine and Ageing Research (IBIMA), Rostock University Medical Center, Rostock, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - M. Hamed
- Institute for Biostatistics and Informatics in Medicine and Ageing Research (IBIMA), Rostock University Medical Center, Rostock, Germany
| | - C. Roolf
- Department of Medicine, Clinic III - Hematology/Oncology/Palliative Care, Rostock University Medical Center, Rostock, Germany
| | - S. Sender
- Department of Medicine, Clinic III - Hematology/Oncology/Palliative Care, Rostock University Medical Center, Rostock, Germany
| | - J. Beck
- Chronix Biomedical GmbH, Göttingen, Germany
| | - E. Schütz
- Chronix Biomedical GmbH, Göttingen, Germany
| | - S. Fischer
- Institute for Biostatistics and Informatics in Medicine and Ageing Research (IBIMA), Rostock University Medical Center, Rostock, Germany
| | - S. Struckmann
- Institute for Biostatistics and Informatics in Medicine and Ageing Research (IBIMA), Rostock University Medical Center, Rostock, Germany
| | - C. Junghanss
- Department of Medicine, Clinic III - Hematology/Oncology/Palliative Care, Rostock University Medical Center, Rostock, Germany
| | - G. Fuellen
- Department of Medicine, Clinic III - Hematology/Oncology/Palliative Care, Rostock University Medical Center, Rostock, Germany
| | - H. Murua Escobar
- Department of Medicine, Clinic III - Hematology/Oncology/Palliative Care, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
18
|
Huynh C, Dingemanse J, Meyer Zu Schwabedissen HE, Sidharta PN. Relevance of the CXCR4/CXCR7-CXCL12 axis and its effect in pathophysiological conditions. Pharmacol Res 2020; 161:105092. [PMID: 32758634 DOI: 10.1016/j.phrs.2020.105092] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/16/2020] [Accepted: 07/19/2020] [Indexed: 02/07/2023]
Abstract
The impact of the C-X-C receptor (CXCR) 7 and its close co-player CXCR4 in different physiological and pathophysiological processes has been extensively investigated within the last decades. Following activation by their shared ligand C-X-C ligand (CXCL) 12, both chemokine receptors can induce various routes of cell signaling and/or scavenge CXCL12 from the extracellular environment. This contributes to organ development and maintenance of homeostasis. Alterations of the CXCR4/CXCR7-CXCL12 axis have been detected in diseases such as cancer, central nervous system and cardiac disorders, and autoimmune diseases. These alterations include changes of the expression pattern, distribution, or downstream effects. The progression of the diseases can be regulated in preclinical models by the use of various modulators suggesting that this axis serves as a promising therapeutic target. It is therefore of great interest to investigate CXCR4/CXCR7/CXCL12 modulators in clinical development, with several CXCR4 and CXCL12 modulators such as plerixafor, ulocuplumab, balixafortide, and olaptesed pegol having already reached this stage. An overview is presented of the most important diseases whose outcomes can be positively or negatively regulated by the CXCR4/CXCR7-CXCL12 axis and summarizes preclinical and clinical data of modulators of that axis. Contrary to CXCR4 and CXCL12 modulators, CXCR7 modulators have, thus far, not been extensively studied. Therefore, more (pre)clinical investigations are needed.
Collapse
Affiliation(s)
- Christine Huynh
- Idorsia Pharmaceuticals Ltd, Department of Clinical Pharmacology, Hegenheimermattweg 91, 4123 Allschwil, Switzerland; Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Jasper Dingemanse
- Idorsia Pharmaceuticals Ltd, Department of Clinical Pharmacology, Hegenheimermattweg 91, 4123 Allschwil, Switzerland
| | | | - Patricia N Sidharta
- Idorsia Pharmaceuticals Ltd, Department of Clinical Pharmacology, Hegenheimermattweg 91, 4123 Allschwil, Switzerland.
| |
Collapse
|
19
|
Kashem MA, Ren X, Li H, Liang B, Li L, Lin F, Plummer FA, Luo M. TILRR Promotes Migration of Immune Cells Through Induction of Soluble Inflammatory Mediators. Front Cell Dev Biol 2020; 8:563. [PMID: 32719797 PMCID: PMC7348050 DOI: 10.3389/fcell.2020.00563] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
TILRR has been identified as an important modulator of inflammatory responses. It is associated with NF-κB activation, and inflammation. Our previous study showed that TILRR significantly increased the expression of many innate immune responsive genes and increased the production of several pro-inflammatory cytokines/chemokines by cervical epithelial cells. In this study, we evaluated the effect of TILRR-induced pro-inflammatory cytokines/chemokines on the migration of immune cells. The effect of culture supernatants of TILRR-overexpressed cervical epithelial cells on the migration of THP-1 monocytes and MOLT-4 T-lymphocytes was evaluated using Transwell assay and a novel microfluidic device. We showed that the culture supernatants of TILRR-overexpressed HeLa cells attracted significantly more THP-1 cells (11–40%, p = 0.0004–0.0373) and MOLT-4 cells (14–17%, p = 0.0010–0.0225) than that of controls. The microfluidic device-recorded image analysis showed that significantly higher amount with longer mean cell migration distance of THP-1 (p < 0.0001–0.0180) and MOLT-4 (p < 0.0001–0.0025) cells was observed toward the supernatants of TILRR-overexpressed cervical epithelial cells compared to that of the controls. Thus, the cytokines/chemokines secreted by the TILRR-overexpressed cervical epithelial cells attracted immune cells, such as monocytes and T cells, and may potentially influence immune cell infiltration in tissues.
Collapse
Affiliation(s)
- Mohammad Abul Kashem
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,JC Wilt Infectious Diseases Research Centre, Winnipeg, MB, Canada.,Department of Microbiology and Veterinary Public Health, Chittagong Veterinary and Animal Sciences University, Chittagong, Bangladesh.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Xiaoou Ren
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB, Canada.,Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB, Canada
| | - Hongzhao Li
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Binhua Liang
- JC Wilt Infectious Diseases Research Centre, Winnipeg, MB, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | - Lin Li
- JC Wilt Infectious Diseases Research Centre, Winnipeg, MB, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Francis Lin
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB, Canada.,Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB, Canada.,Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Francis A Plummer
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Ma Luo
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,JC Wilt Infectious Diseases Research Centre, Winnipeg, MB, Canada.,National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| |
Collapse
|
20
|
Groblewska M, Litman-Zawadzka A, Mroczko B. The Role of Selected Chemokines and Their Receptors in the Development of Gliomas. Int J Mol Sci 2020; 21:ijms21103704. [PMID: 32456359 PMCID: PMC7279280 DOI: 10.3390/ijms21103704] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Among heterogeneous primary tumors of the central nervous system (CNS), gliomas are the most frequent type, with glioblastoma multiforme (GBM) characterized with the worst prognosis. In their development, certain chemokine/receptor axes play important roles and promote proliferation, survival, metastasis, and neoangiogenesis. However, little is known about the significance of atypical receptors for chemokines (ACKRs) in these tumors. The objective of the study was to present the role of chemokines and their conventional and atypical receptors in CNS tumors. Therefore, we performed a thorough search for literature concerning our investigation via the PubMed database. We describe biological functions of chemokines/chemokine receptors from various groups and their significance in carcinogenesis, cancer-related inflammation, neo-angiogenesis, tumor growth, and metastasis. Furthermore, we discuss the role of chemokines in glioma development, with particular regard to their function in the transition from low-grade to high-grade tumors and angiogenic switch. We also depict various chemokine/receptor axes, such as CXCL8-CXCR1/2, CXCL12-CXCR4, CXCL16-CXCR6, CX3CL1-CX3CR1, CCL2-CCR2, and CCL5-CCR5 of special importance in gliomas, as well as atypical chemokine receptors ACKR1-4, CCRL2, and PITPMN3. Additionally, the diagnostic significance and usefulness of the measurement of some chemokines and their receptors in the blood and cerebrospinal fluid (CSF) of glioma patients is also presented.
Collapse
Affiliation(s)
- Magdalena Groblewska
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland;
| | - Ala Litman-Zawadzka
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland;
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland;
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland;
- Correspondence: ; Tel.: +48-85-831-8785
| |
Collapse
|
21
|
Capella Roca B, Doolan P, Barron N, O'Neill F, Clynes M. Altered gene expression in CHO cells following polyamine starvation. Biotechnol Lett 2020; 42:927-936. [PMID: 32078082 DOI: 10.1007/s10529-020-02841-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/15/2020] [Indexed: 11/27/2022]
Abstract
AIM To investigate the impact of polyamine deprivation on the transcriptome of CHO cells RESULTS: Polyamines play a central but poorly-understood role in cell proliferation. Most studies to date have utilised chemical inhibitors to probe polyamine function. Here we exploit the fact that CHO cells grown in serum-free medium have an absolute requirement for putrescine supplementation due to their deficiency in activity of the enzyme arginase. A gene expression microarray (Affymetrix) analysis of CHO-K1 cells starved of polyamines for 3 days showed that cessation of growth, associated with increased G1/S transition and inhibition of M/G1 transition was accompanied by increased mRNA levels of mitotic complex checkpoint genes (Mad2l1, Tkk, Bub1b) and in the transition of G1- to S-phase (such as Skp2 and Tfdp1). mRNAs associated with DNA homologous recombination and repair (including Fanconi's anaemia-related genes) and with RNA splicing were consistently increased. Alterations in mRNA levels for genes related to protein processing in the ER, to ER stress, and to p53-related and apoptosis pathways were also observed. mRNAs showing highest levels of fold-change included several which code for membrane-localised proteins and receptors (Thbs1, Tfrc1, Ackr3, Extl1). CONCLUSIONS Growth-arrest induced by polyamine deprivation was associated with significant alterations in levels of mRNAs associated with cell cycle progression, DNA repair, RNA splicing, ER trafficking and membrane signalling as well as p53 and apoptosis-related pathways.
Collapse
Affiliation(s)
- Berta Capella Roca
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
- SSPC-SFI Centre for Pharmaceuticals, Dublin City University, Dublin 9, Ireland
| | - Padraig Doolan
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Niall Barron
- National Institute for Bioprocessing Research and Training, University College Dublin, Dublin, Ireland
| | - Fiona O'Neill
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Martin Clynes
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
- SSPC-SFI Centre for Pharmaceuticals, Dublin City University, Dublin 9, Ireland.
| |
Collapse
|
22
|
Montaño A, Ordoñez JL, Alonso-Pérez V, Hernández-Sánchez J, Santos S, González T, Benito R, García-Tuñón I, Hernández-Rivas JM. ETV6/ RUNX1 Fusion Gene Abrogation Decreases the Oncogenicity of Tumour Cells in a Preclinical Model of Acute Lymphoblastic Leukaemia. Cells 2020; 9:E215. [PMID: 31952221 PMCID: PMC7017301 DOI: 10.3390/cells9010215] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The t(12;21)(p13;q22), which fuses ETV6 and RUNX1 genes, is the most common genetic abnormality in children with B-cell precursor acute lymphoblastic leukaemia. The implication of the fusion protein in leukemogenesis seems to be clear. However, its role in the maintenance of the disease continues to be controversial. METHODS Generation of an in vitroETV6/RUNX1 knock out model using the CRISPR/Cas9 gene editing system. Functional characterization by RNA sequencing, proliferation assays, apoptosis and pharmacologic studies, and generation of edited-cell xenograft model. RESULTS The expression of ETV6/RUNX1 fusion gene was completely eliminated, thus generating a powerful model on which to study the role of the fusion gene in leukemic cells. The loss of fusion gene expression led to the deregulation of biological processes affecting survival such as apoptosis resistance and cell proliferation capacity. Tumour cells showed higher levels of apoptosis, lower proliferation rate and a greater sensitivity to PI3K inhibitors in vitro along as a decrease in tumour growth in xenografts models after ETV6/RUNX1 fusion gene abrogation. CONCLUSIONS ETV6/RUNX1 fusion protein seems to play an important role in the maintenance of the leukemic phenotype and could thus become a potential therapeutic target.
Collapse
Affiliation(s)
- Adrián Montaño
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
| | - Jose Luis Ordoñez
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
- Department of Biochemistry and Molecular Biology, University of Salamanca, Campus Unamuno s/n, 37007 Salamanca, Spain
| | - Verónica Alonso-Pérez
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
| | - Jesús Hernández-Sánchez
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
| | - Sandra Santos
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
| | - Teresa González
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
- Department of Hematology, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Rocío Benito
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
| | - Ignacio García-Tuñón
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
| | - Jesús María Hernández-Rivas
- IBSAL, IBMCC, Cancer Research Center, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain; (A.M.); (J.L.O.); (V.A.-P.); (J.H.-S.); (S.S.); (T.G.); (R.B.)
- Department of Hematology, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Department of Medicine, Universidad de Salamanca and CIBERONC, 37007 Salamanca, Spain
| |
Collapse
|
23
|
Koenen J, Bachelerie F, Balabanian K, Schlecht-Louf G, Gallego C. Atypical Chemokine Receptor 3 (ACKR3): A Comprehensive Overview of its Expression and Potential Roles in the Immune System. Mol Pharmacol 2019; 96:809-818. [PMID: 31040166 DOI: 10.1124/mol.118.115329] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/24/2019] [Indexed: 12/24/2022] Open
Abstract
Atypical chemokine receptor 3 (ACKR3), previously known as C-X-C chemokine receptor type 7 (CXCR7), has emerged as a key player in several biologic processes, particularly during development. Its CXCL11 and CXCL12 scavenging activity and atypical signaling properties, together with a new array of other nonchemokine ligands, have established ACKR3 as a main regulator of physiologic processes at steady state and during inflammation. Here, we present a comprehensive review of ACKR3 expression in mammalian tissues in search of a possible connection with the receptor function. Besides the reported roles of ACKR3 during development, we discuss the potential contribution of ACKR3 to the function of the immune system, focusing on the myeloid lineage.
Collapse
Affiliation(s)
- Joyce Koenen
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Université Paris-Sud and Université Paris-Saclay, Clamart, France (J.K., F.B., K.B., G.S.-L., C.G.) and Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.K.)
| | - Françoise Bachelerie
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Université Paris-Sud and Université Paris-Saclay, Clamart, France (J.K., F.B., K.B., G.S.-L., C.G.) and Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.K.)
| | - Karl Balabanian
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Université Paris-Sud and Université Paris-Saclay, Clamart, France (J.K., F.B., K.B., G.S.-L., C.G.) and Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.K.)
| | - Géraldine Schlecht-Louf
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Université Paris-Sud and Université Paris-Saclay, Clamart, France (J.K., F.B., K.B., G.S.-L., C.G.) and Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.K.)
| | - Carmen Gallego
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Université Paris-Sud and Université Paris-Saclay, Clamart, France (J.K., F.B., K.B., G.S.-L., C.G.) and Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.K.)
| |
Collapse
|
24
|
Bobkov V, Arimont M, Zarca A, De Groof TWM, van der Woning B, de Haard H, Smit MJ. Antibodies Targeting Chemokine Receptors CXCR4 and ACKR3. Mol Pharmacol 2019; 96:753-764. [PMID: 31481460 DOI: 10.1124/mol.119.116954] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/03/2019] [Indexed: 12/19/2022] Open
Abstract
Dysregulation of the chemokine system is implicated in a number of autoimmune and inflammatory diseases, as well as cancer. Modulation of chemokine receptor function is a very promising approach for therapeutic intervention. Despite interest from academic groups and pharmaceutical companies, there are currently few approved medicines targeting chemokine receptors. Monoclonal antibodies (mAbs) and antibody-based molecules have been successfully applied in the clinical therapy of cancer and represent a potential new class of therapeutics targeting chemokine receptors belonging to the class of G protein-coupled receptors (GPCRs). Besides conventional mAbs, single-domain antibodies and antibody scaffolds are also gaining attention as promising therapeutics. In this review, we provide an extensive overview of mAbs, single-domain antibodies, and other antibody fragments targeting CXCR4 and ACKR3, formerly referred to as CXCR7. We discuss their unique properties and advantages over small-molecule compounds, and also refer to the molecules in preclinical and clinical development. We focus on single-domain antibodies and scaffolds and their utilization in GPCR research. Additionally, structural analysis of antibody binding to CXCR4 is discussed. SIGNIFICANCE STATEMENT: Modulating the function of GPCRs, and particularly chemokine receptors, draws high interest. A comprehensive review is provided for monoclonal antibodies, antibody fragments, and variants directed at CXCR4 and ACKR3. Their advantageous functional properties, versatile applications as research tools, and use in the clinic are discussed.
Collapse
Affiliation(s)
- Vladimir Bobkov
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (V.B., M.A., A.Z., T.W.M.D.G., M.J.S.); and argenx BVBA, Zwijnaarde, Belgium (V.B., B.W., H.H.)
| | - Marta Arimont
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (V.B., M.A., A.Z., T.W.M.D.G., M.J.S.); and argenx BVBA, Zwijnaarde, Belgium (V.B., B.W., H.H.)
| | - Aurélien Zarca
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (V.B., M.A., A.Z., T.W.M.D.G., M.J.S.); and argenx BVBA, Zwijnaarde, Belgium (V.B., B.W., H.H.)
| | - Timo W M De Groof
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (V.B., M.A., A.Z., T.W.M.D.G., M.J.S.); and argenx BVBA, Zwijnaarde, Belgium (V.B., B.W., H.H.)
| | - Bas van der Woning
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (V.B., M.A., A.Z., T.W.M.D.G., M.J.S.); and argenx BVBA, Zwijnaarde, Belgium (V.B., B.W., H.H.)
| | - Hans de Haard
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (V.B., M.A., A.Z., T.W.M.D.G., M.J.S.); and argenx BVBA, Zwijnaarde, Belgium (V.B., B.W., H.H.)
| | - Martine J Smit
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (V.B., M.A., A.Z., T.W.M.D.G., M.J.S.); and argenx BVBA, Zwijnaarde, Belgium (V.B., B.W., H.H.)
| |
Collapse
|
25
|
Notch/CXCR4 Partnership in Acute Lymphoblastic Leukemia Progression. J Immunol Res 2019; 2019:5601396. [PMID: 31346528 PMCID: PMC6620846 DOI: 10.1155/2019/5601396] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 05/21/2019] [Accepted: 06/12/2019] [Indexed: 02/08/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common cancer among children. Recent advances in chemotherapy have made ALL a curable hematological malignancy. In children, there is 25% chance of disease relapse, typically in the central nervous system. While in adults, there is a higher chance of relapse. ALL may affect B-cell or T-cell lineages. Different genetic alterations characterize the two ALL forms. Deregulated Notch, either Notch1 or Notch3, and CXCR4 receptor signaling are involved in ALL disease development and progression. By analyzing their relevant roles in the pathogenesis of the two ALL forms, new molecular mechanisms able to modulate cancer cell invasion may be visualized. Notably, the partnership between Notch and CXCR4 may have considerable implications in understanding the complexity of T- and B-ALL. These two receptor pathways intersect other critical signals in the proliferative, differentiation, and metabolic programs of lymphocyte transformation. Also, the identification of the crosstalks in leukemia-stroma interaction within the tumor microenvironment may unveil new targetable mechanisms in disease relapse. Further studies are required to identify new challenges and opportunities to develop more selective and safer therapeutic strategies in ALL progression, possibly contributing to improve conventional hematological cancer therapy.
Collapse
|
26
|
Abstract
Although miR-101 is involved in the development and progression of T-cell acute lymphoblastic leukemia (T-ALL), the underlying molecular mechanisms remain unclear. In this article, we report that miR-101 expression was inversely correlated with CX chemokine receptor 7 (CXCR7) level in T-ALL. Introducing miR-101 inhibited T-ALL cell proliferation and invasion in vitro and suppressed tumor growth and lung metastasis in vivo. CXCR7 was identified as a direct target of miR-101. The inhibitory effects of miR-101 were mimicked and counteracted by CXCR7 depletion and overexpression, respectively. Mechanistically, miR-101 targets CXCR7/STAT3 axis to reduce T-ALL growth and metastasis. Overall, these findings implied the potential application of miR-101 and CXCR7 in T-ALL treatment.
Collapse
Affiliation(s)
- Xue-Yi Yang
- Life Science College, Luoyang Normal University, Luoyang, Henan, P.R. China
| | - Ye Sheng
- Life Science College, Luoyang Normal University, Luoyang, Henan, P.R. China
| |
Collapse
|
27
|
Redondo-Muñoz J, García-Pardo A, Teixidó J. Molecular Players in Hematologic Tumor Cell Trafficking. Front Immunol 2019; 10:156. [PMID: 30787933 PMCID: PMC6372527 DOI: 10.3389/fimmu.2019.00156] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/17/2019] [Indexed: 12/20/2022] Open
Abstract
The trafficking of neoplastic cells represents a key process that contributes to progression of hematologic malignancies. Diapedesis of neoplastic cells across endothelium and perivascular cells is facilitated by adhesion molecules and chemokines, which act in concert to tightly regulate directional motility. Intravital microscopy provides spatio-temporal views of neoplastic cell trafficking, and is crucial for testing and developing therapies against hematologic cancers. Multiple myeloma (MM), chronic lymphocytic leukemia (CLL), and acute lymphoblastic leukemia (ALL) are hematologic malignancies characterized by continuous neoplastic cell trafficking during disease progression. A common feature of these neoplasias is the homing and infiltration of blood cancer cells into the bone marrow (BM), which favors growth and survival of the malignant cells. MM cells traffic between different BM niches and egress from BM at late disease stages. Besides the BM, CLL cells commonly home to lymph nodes (LNs) and spleen. Likewise, ALL cells also infiltrate extramedullary organs, such as the central nervous system, spleen, liver, and testicles. The α4β1 integrin and the chemokine receptor CXCR4 are key molecules for MM, ALL, and CLL cell trafficking into and out of the BM. In addition, the chemokine receptor CCR7 controls CLL cell homing to LNs, and CXCR4, CCR7, and CXCR3 contribute to ALL cell migration across endothelia and the blood brain barrier. Some of these receptors are used as diagnostic markers for relapse and survival in ALL patients, and their level of expression allows clinicians to choose the appropriate treatments. In CLL, elevated α4β1 expression is an established adverse prognostic marker, reinforcing its role in the disease expansion. Combining current chemotherapies with inhibitors of malignant cell trafficking could represent a useful therapy against these neoplasias. Moreover, immunotherapy using humanized antibodies, CAR-T cells, or immune check-point inhibitors together with agents targeting the migration of tumor cells could also restrict their survival. In this review, we provide a view of the molecular players that regulate the trafficking of neoplastic cells during development and progression of MM, CLL, and ALL, together with current therapies that target the malignant cells.
Collapse
Affiliation(s)
- Javier Redondo-Muñoz
- Department of Immunology, Ophthalmology and ERL, Hospital 12 de Octubre Health Research Institute (imas12), School of Medicine, Complutense University, Madrid, Spain.,Manchester Collaborative Centre for Inflammation Research, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Angeles García-Pardo
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - Joaquin Teixidó
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| |
Collapse
|
28
|
Borroni EM, Savino B, Bonecchi R, Locati M. Chemokines sound the alarmin: The role of atypical chemokine in inflammation and cancer. Semin Immunol 2018; 38:63-71. [DOI: 10.1016/j.smim.2018.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/08/2018] [Indexed: 12/17/2022]
|
29
|
Moreno MJ, Gallardo A, Novelli S, Mozos A, Aragó M, Pavón MÁ, Céspedes MV, Pallarès V, Falgàs A, Alcoceba M, Blanco O, Gonzalez-Díaz M, Sierra J, Mangues R, Casanova I. CXCR7 expression in diffuse large B-cell lymphoma identifies a subgroup of CXCR4+ patients with good prognosis. PLoS One 2018; 13:e0198789. [PMID: 29920526 PMCID: PMC6007902 DOI: 10.1371/journal.pone.0198789] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 04/14/2018] [Indexed: 01/10/2023] Open
Abstract
The CXCR4/CXCL12 axis has been extensively associated with different types of cancer correlating with higher aggressiveness and metastasis. In diffuse large B-cell lymphoma (DLBCL), the expression of the chemokine receptor CXCR4 is involved in the dissemination of malignant B cells and is a marker of poor prognosis. CXCR7 is a chemokine receptor that binds to the same ligand as CXCR4 and regulates de CXCR4-CXCL12 axis. These findings together with the report of CXCR7 prognostic value in several tumor types, led us to evaluate the expression of CXCR7 in diffuse large B-cell lymphoma biopsies. Here, we describe that CXCR7 receptor is an independent prognostic factor that associates with good clinical outcome. Moreover, the expression of CXCR7 associates with increased survival in CXCR4+ but not in CXCR4- DLBCL patients. Thus, the combined immunohistochemical evaluation of both CXCR7 and CXCR4 expression in DLBCL biopsies may improve their prognostic value as single markers. Finally, we show that CXCR7 overexpression in vitro is able to diminish DLBCL cell survival and increase their sensitivity to antitumor drugs. Hence, further studies on the CXCR7 receptor may establish its role in DLBCL and the molecular mechanisms that modulate CXCR4 activity.
Collapse
MESH Headings
- Adult
- Aged
- Biomarkers, Tumor
- Biopsy
- Cell Line, Tumor
- Chemokine CXCL12/physiology
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Kaplan-Meier Estimate
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Male
- Middle Aged
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Prognosis
- Proportional Hazards Models
- Receptors, CXCR/biosynthesis
- Receptors, CXCR/genetics
- Receptors, CXCR/physiology
- Receptors, CXCR4/analysis
Collapse
Affiliation(s)
- María José Moreno
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
| | - Alberto Gallardo
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Silvana Novelli
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ana Mozos
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Marc Aragó
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Miguel Ángel Pavón
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
| | - María Virtudes Céspedes
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
| | - Víctor Pallarès
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Aïda Falgàs
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Miguel Alcoceba
- Department of Hematology and Pathology, IBSAL-University Hospital, Center for Cancer Research-IBMCC (USAL-CSIC), Salamanca, Spain
- CIBER in Oncology (CIBER-ONC), Madrid, Spain
| | - Oscar Blanco
- Department of Hematology and Pathology, IBSAL-University Hospital, Center for Cancer Research-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Marcos Gonzalez-Díaz
- Department of Hematology and Pathology, IBSAL-University Hospital, Center for Cancer Research-IBMCC (USAL-CSIC), Salamanca, Spain
- CIBER in Oncology (CIBER-ONC), Madrid, Spain
| | - Jorge Sierra
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Josep Carreras Research Institute, Barcelona, Spain
| | - Ramon Mangues
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
- Josep Carreras Research Institute, Barcelona, Spain
- * E-mail:
| | - Isolda Casanova
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
- Josep Carreras Research Institute, Barcelona, Spain
| |
Collapse
|
30
|
Piovan E, Tosello V, Amadori A, Zanovello P. Chemotactic Cues for NOTCH1-Dependent Leukemia. Front Immunol 2018; 9:633. [PMID: 29666622 PMCID: PMC5891592 DOI: 10.3389/fimmu.2018.00633] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/14/2018] [Indexed: 12/12/2022] Open
Abstract
The NOTCH signaling pathway is a conserved signaling cascade that regulates many aspects of development and homeostasis in multiple organ systems. Aberrant activity of this signaling pathway is linked to the initiation and progression of several hematological malignancies, exemplified by T-cell acute lymphoblastic leukemia (T-ALL). Interestingly, frequent non-mutational activation of NOTCH1 signaling has recently been demonstrated in B-cell chronic lymphocytic leukemia (B-CLL), significantly extending the pathogenic significance of this pathway in B-CLL. Leukemia patients often present with high-blood cell counts, diffuse disease with infiltration of the bone marrow, secondary lymphoid organs, and diffusion to the central nervous system (CNS). Chemokines are chemotactic cytokines that regulate migration of cells between tissues and the positioning and interactions of cells within tissue. Homeostatic chemokines and their receptors have been implicated in regulating organ-specific infiltration, but may also directly and indirectly modulate tumor growth. Recently, oncogenic NOTCH1 has been shown to regulate infiltration of leukemic cells into the CNS hijacking the CC-chemokine ligand 19/CC-chemokine receptor 7 chemokine axis. In addition, a crucial role for the homing receptor axis CXC-chemokine ligand 12/CXC-chemokine receptor 4 has been demonstrated in leukemia maintenance and progression. Moreover, the CCL25/CCR9 axis has been implicated in the homing of leukemic cells into the gut, particularly in the presence of phosphatase and tensin homolog tumor suppressor loss. In this review, we summarize the latest developments regarding the role of NOTCH signaling in regulating the chemotactic microenvironmental cues involved in the generation and progression of T-ALL and compare these findings to B-CLL.
Collapse
Affiliation(s)
- Erich Piovan
- UOC Immunologia e Diagnostica Molecolare Oncologica, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy.,Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Università di Padova, Padova, Italy
| | - Valeria Tosello
- UOC Immunologia e Diagnostica Molecolare Oncologica, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Alberto Amadori
- UOC Immunologia e Diagnostica Molecolare Oncologica, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy.,Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Università di Padova, Padova, Italy
| | - Paola Zanovello
- UOC Immunologia e Diagnostica Molecolare Oncologica, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy.,Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Università di Padova, Padova, Italy
| |
Collapse
|
31
|
Melo RDCC, Ferro KPV, Duarte ADSS, Olalla Saad ST. CXCR7 participates in CXCL12-mediated migration and homing of leukemic and normal hematopoietic cells. Stem Cell Res Ther 2018; 9:34. [PMID: 29433559 PMCID: PMC5810108 DOI: 10.1186/s13287-017-0765-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 12/26/2017] [Indexed: 12/30/2022] Open
Abstract
CXCR4 was the first receptor identified for CXCL12, but a second receptor, CXCR7, has also been described and its function in hematopoietic cells remains unknown. By inhibition of CXCR4 and/or CXCR7, we showed that CXCR7 participates in normal CD34+ and U937 cell migration and prevents downregulation of CXCR4 by CXCL12 stimulation. In addition, CXCR7 contributes to homing of acute myeloid leukemia and normal progenitor cells to the bone marrow and spleen of NOD/SCID mice. In summary, this study shows an essential role of CXCR7, together with CXCR4, in the control of normal and malignant hematopoietic cell migration and homing induced by CXCL12.
Collapse
Affiliation(s)
- Rita de Cassia Carvalho Melo
- Hematology and Transfusion Medicine Center, University of Campinas/Hemocentro UNICAMP, Campinas, São Paulo, Brazil
| | - Karla Priscila Viera Ferro
- Hematology and Transfusion Medicine Center, University of Campinas/Hemocentro UNICAMP, Campinas, São Paulo, Brazil
| | | | - Sara Teresinha Olalla Saad
- Hematology and Transfusion Medicine Center, University of Campinas/Hemocentro UNICAMP, Campinas, São Paulo, Brazil.
| |
Collapse
|
32
|
Shi A, Shi H, Dong L, Xu S, Jia M, Guo X, Wang T. CXCR7 as a chemokine receptor for SDF-1 promotes gastric cancer progression via MAPK pathways. Scand J Gastroenterol 2017; 52:745-753. [PMID: 28281844 DOI: 10.1080/00365521.2017.1300681] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE As the alternate receptor for stromal cell-derived factor-1 (SDF-1) except CXCR4, CXCR7 has been shown to be involved in the progression of some malignancies. However, the role of SDF-1/CXCR7 in gastric cancer (GC) remains unclear. MATERIALS AND METHODS CXCR7 expression was examined in 83 human GC tissues and adjacent non-cancer tissues (ANCTs) by immunohistochemistry, in three human GC cell lines (MGC-803, BGC-823 and SGC-7901) by reverse transcription-PCR and western blot. CXCR7 was stably knocked down via lentiviral vectors. The cells proliferation was evaluated using CCK-8 and colony formation assay; MAPK pathways (ERK1/2, p38 and SAPK/JNK) were detected using western blot. Besides, the xenograft model of nude mice for GC growth was performed. RESULTS CXCR7 expression in GC tissues was significantly higher than that in ANCTs and associated with tumor size, TNM stage and lymph node metastasis. CXCR7 and CXCR4 were both detectable in three GC cell lines and SGC-7901 cells expressed CXCR7 the most abundantly. SDF-1 promoted the proliferation of SGC-7901 cells, the phosphorylation of ERK1/2, p38 and CXCR7 knockdown distinctly reversed these changes; the proliferation stimulated with SDF-1 was attenuated by U0126 (MEK1/2 inhibitor). Furthermore, CXCR7 knockdown inhibited the growth of GC subcutaneous xenografts and decreased the microvessel density and VEGF expression in vivo. CONCLUSION CXCR7 was identified as a novel promoter in GC initiation and progression.
Collapse
Affiliation(s)
- Ameng Shi
- a Department of Gastroenterology , The Second Affiliated Hospital of Xi'an Jiaotong University , Shaanxi , China
| | - Haitao Shi
- a Department of Gastroenterology , The Second Affiliated Hospital of Xi'an Jiaotong University , Shaanxi , China
| | - Lei Dong
- a Department of Gastroenterology , The Second Affiliated Hospital of Xi'an Jiaotong University , Shaanxi , China
| | - Shaoxian Xu
- a Department of Gastroenterology , The Second Affiliated Hospital of Xi'an Jiaotong University , Shaanxi , China
| | - Miao Jia
- a Department of Gastroenterology , The Second Affiliated Hospital of Xi'an Jiaotong University , Shaanxi , China
| | - Xiaoyan Guo
- a Department of Gastroenterology , The Second Affiliated Hospital of Xi'an Jiaotong University , Shaanxi , China
| | - Ting Wang
- a Department of Gastroenterology , The Second Affiliated Hospital of Xi'an Jiaotong University , Shaanxi , China
| |
Collapse
|
33
|
Umezawa Y, Akiyama H, Okada K, Ishida S, Nogami A, Oshikawa G, Kurosu T, Miura O. Molecular mechanisms for enhancement of stromal cell-derived factor 1-induced chemotaxis by platelet endothelial cell adhesion molecule 1 (PECAM-1). J Biol Chem 2017; 292:19639-19655. [PMID: 28974577 DOI: 10.1074/jbc.m117.779603] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 09/26/2017] [Indexed: 01/16/2023] Open
Abstract
Platelet endothelial cell adhesion molecule 1 (PECAM-1) is a cell adhesion protein involved in the regulation of cell adhesion and migration. Interestingly, several PECAM-1-deficient hematopoietic cells exhibit impaired chemotactic responses to stromal cell-derived factor 1 (SDF-1), a chemokine essential for B lymphopoiesis and bone marrow myelopoiesis. However, whether PECAM-1 is involved in SDF-1-regulated chemotaxis is unknown. We report here that SDF-1 induces tyrosine phosphorylation of PECAM-1 at its immunoreceptor tyrosine-based inhibition motifs in several hematopoietic cell lines via the Src family kinase Lyn, Bruton's tyrosine kinase, and JAK2 and that inhibition of these kinases reduced chemotaxis. Overexpression and knockdown of PECAM-1 enhanced and down-regulated, respectively, SDF-1-induced Gαi-dependent activation of the PI3K/Akt/mTORC1 pathway and small GTPase Rap1 in hematopoietic 32Dcl3 cells, and these changes in activation correlated with chemotaxis. Furthermore, pharmacological or genetic inhibition of the PI3K/Akt/mTORC1 pathway or Rap1, respectively, revealed that these pathways are independently activated and required for SDF-1-induced chemotaxis. When coexpressed in 293T cells, PECAM-1 physically associated with the SDF-1 receptor CXCR4. Moreover, PECAM-1 overexpression and knockdown reduced and enhanced SDF-1-induced endocytosis of CXCR4, respectively. Furthermore, when expressed in 32Dcl3 cells, an endocytosis-defective CXCR4 mutant, CXCR4-S324A/S325A, could activate the PI3K/Akt/mTORC1 pathway as well as Rap1 and induce chemotaxis in a manner similar to PECAM-1 overexpression. These findings suggest that PECAM-1 enhances SDF-1-induced chemotaxis by augmenting and prolonging activation of the PI3K/Akt/mTORC1 pathway and Rap1 and that PECAM-1, at least partly, exerts its activity by inhibiting SDF-1-induced internalization of CXCR4.
Collapse
Affiliation(s)
- Yoshihiro Umezawa
- From the Department of Hematology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyoku, Tokyo 113-8519, Japan
| | - Hiroki Akiyama
- From the Department of Hematology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyoku, Tokyo 113-8519, Japan
| | - Keigo Okada
- From the Department of Hematology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyoku, Tokyo 113-8519, Japan
| | - Shinya Ishida
- From the Department of Hematology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyoku, Tokyo 113-8519, Japan
| | - Ayako Nogami
- From the Department of Hematology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyoku, Tokyo 113-8519, Japan
| | - Gaku Oshikawa
- From the Department of Hematology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyoku, Tokyo 113-8519, Japan
| | - Tetsuya Kurosu
- From the Department of Hematology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyoku, Tokyo 113-8519, Japan
| | - Osamu Miura
- From the Department of Hematology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyoku, Tokyo 113-8519, Japan
| |
Collapse
|
34
|
Bongiovanni D, Saccomani V, Piovan E. Aberrant Signaling Pathways in T-Cell Acute Lymphoblastic Leukemia. Int J Mol Sci 2017; 18:ijms18091904. [PMID: 28872614 PMCID: PMC5618553 DOI: 10.3390/ijms18091904] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 08/30/2017] [Accepted: 09/01/2017] [Indexed: 12/12/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive disease caused by the malignant transformation of immature progenitors primed towards T-cell development. Clinically, T-ALL patients present with diffuse infiltration of the bone marrow by immature T-cell blasts high blood cell counts, mediastinal involvement, and diffusion to the central nervous system. In the past decade, the genomic landscape of T-ALL has been the target of intense research. The identification of specific genomic alterations has contributed to identify strong oncogenic drivers and signaling pathways regulating leukemia growth. Notwithstanding, T-ALL patients are still treated with high-dose multiagent chemotherapy, potentially exposing these patients to considerable acute and long-term side effects. This review summarizes recent advances in our understanding of the signaling pathways relevant for the pathogenesis of T-ALL and the opportunities offered for targeted therapy.
Collapse
Affiliation(s)
- Deborah Bongiovanni
- Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Universita' di Padova, Padova 35128, Italy.
| | - Valentina Saccomani
- Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Universita' di Padova, Padova 35128, Italy.
| | - Erich Piovan
- Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Universita' di Padova, Padova 35128, Italy.
- UOC Immunologia e Diagnostica Molecolare Oncologica, Istituto Oncologico Veneto IOV-IRCCS, Padova 35128, Italy.
| |
Collapse
|
35
|
The role of G protein-coupled receptors in lymphoid malignancies. Cell Signal 2017; 39:95-107. [PMID: 28802842 DOI: 10.1016/j.cellsig.2017.08.002] [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: 05/25/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 12/20/2022]
Abstract
B cell lymphoma consists of multiple individual diseases arising throughout the lifespan of B cell development. From pro-B cells in the bone marrow, through circulating mature memory B cells, each stage of B cell development is prone to oncogenic mutation and transformation, which can lead to a corresponding lymphoma. Therapies designed against individual types of lymphoma often target features that differ between malignant cells and the corresponding normal cells from which they arise. These genetic changes between tumor and normal cells can include oncogene activation, tumor suppressor gene repression and modified cell surface receptor expression. G protein-coupled receptors (GPCRs) are an important class of cell surface receptors that represent an ideal target for lymphoma therapeutics. GPCRs bind a wide range of ligands to relay extracellular signals through G protein-mediated signaling cascades. Each lymphoma subgroup expresses a unique pattern of GPCRs and efforts are underway to fully characterize these patterns at the genetic level. Aberrations such as overexpression, deletion and mutation of GPCRs have been characterized as having causative roles in lymphoma and such studies describing GPCRs in B cell lymphomas are summarized here.
Collapse
|
36
|
Puddinu V, Casella S, Radice E, Thelen S, Dirnhofer S, Bertoni F, Thelen M. ACKR3 expression on diffuse large B cell lymphoma is required for tumor spreading and tissue infiltration. Oncotarget 2017; 8:85068-85084. [PMID: 29156704 PMCID: PMC5689594 DOI: 10.18632/oncotarget.18844] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/05/2017] [Indexed: 12/30/2022] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is the most frequent lymphoma accounting for more than the 30% of the cases. Involvement of extranodal sites, such as bone marrow and central nervous system, is associated with poor prognosis. A contribution of the chemokine system in these processes is assumed as it is known as a critical regulator of the metastatic process in cancer. The atypical chemokine receptor 3 (ACKR3), which does not couple to G-proteins and does not mediate cell migration, acts as a scavenger for CXCL11 and CXCL12, interfering with the tumor homing CXCL12/CXCR4 axis. Here, functional expression of ACKR3 in DLBCL cells was necessary for colonization of the draining lymph node in an in vivo subcutaneous lymphoma model. Moreover, in a disseminated in vivo lymphoma model, ACKR3 expression was required for bone marrow and brain invasion and local tumor growth. The present data unveil ACKR3 as potential therapeutic target for the control of tumor dissemination in DLBCL.
Collapse
Affiliation(s)
- Viola Puddinu
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Sabrina Casella
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Egle Radice
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Sylvia Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Stefan Dirnhofer
- Institute of Pathology, University Hospital, University of Basel, Basel, Switzerland
| | | | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| |
Collapse
|
37
|
Oliveira ML, Akkapeddi P, Alcobia I, Almeida AR, Cardoso BA, Fragoso R, Serafim TL, Barata JT. From the outside, from within: Biological and therapeutic relevance of signal transduction in T-cell acute lymphoblastic leukemia. Cell Signal 2017. [PMID: 28645565 DOI: 10.1016/j.cellsig.2017.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological cancer that arises from clonal expansion of transformed T-cell precursors. In this review we summarize the current knowledge on the external stimuli and cell-intrinsic lesions that drive aberrant activation of pivotal, pro-tumoral intracellular signaling pathways in T-cell precursors, driving transformation, leukemia expansion, spread or resistance to therapy. In addition to their pathophysiological relevance, receptors and kinases involved in signal transduction are often attractive candidates for targeted drug development. As such, we discuss also the potential of T-ALL signaling players as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Mariana L Oliveira
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Padma Akkapeddi
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Isabel Alcobia
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Afonso R Almeida
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Bruno A Cardoso
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Rita Fragoso
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Teresa L Serafim
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - João T Barata
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal.
| |
Collapse
|
38
|
Ngamsri KC, Müller A, Bösmüller H, Gamper-Tsigaras J, Reutershan J, Konrad FM. The Pivotal Role of CXCR7 in Stabilization of the Pulmonary Epithelial Barrier in Acute Pulmonary Inflammation. THE JOURNAL OF IMMUNOLOGY 2017; 198:2403-2413. [PMID: 28188248 DOI: 10.4049/jimmunol.1601682] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/12/2017] [Indexed: 11/19/2022]
Abstract
Acute pulmonary inflammation is still a frightening complication in intensive care units and has a high mortality. Specific treatment is not available, and many details of the pathomechanism remain unclear. The recently discovered chemokine receptor CXCR7 and its ligand stromal cell-derived factor (SDF)-1 are known to be involved in inflammation. We chose to investigate the detailed role of CXCR7 in a murine model of LPS inhalation. Inflammation increased pulmonary expression of CXCR7, and the receptor was predominantly expressed on pulmonary epithelium and on polymorphonuclear neutrophil (PMNs) after transepithelial migration into the alveolar space. Specific inhibition of CXCR7 reduced transepithelial PMN migration by affecting the expression of adhesion molecules. CXCR7 antagonism reduced the most potent PMN chemoattractants CXCL1 and CXCL2/3. After inhibiting CXCR7, NF-κB phosphorylation was reduced in lungs of mice, tight junction formation increased, and protein concentration in the bronchoalveolar lavage diminished, showing the impact of CXCR7 on stabilizing microvascular permeability. In vitro studies with human cells confirmed the pivotal role of CXCR7 in pulmonary epithelium. Immunofluorescence of human lungs confirmed our in vivo data and showed an increase of the expression of CXCR7 in pulmonary epithelium. Highlighting the clinical potential of CXCR7 antagonism, nebulization of the agent before and after the inflammation showed impressive anti-inflammatory effects. Additional CXCR7 inhibition potentiated the effect of SDF-1 antagonism, most probably by downregulating SDF-1 and the second receptor of the chemokine (CXCR4) expression. In conclusion, our data identified the pivotal role of the receptor CXCR7 in pulmonary inflammation with a predominant effect on the pulmonary epithelium and PMNs.
Collapse
Affiliation(s)
- Kristian-Christos Ngamsri
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tübingen, 72076 Tübingen, Germany
| | - Anika Müller
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tübingen, 72076 Tübingen, Germany
| | - Hans Bösmüller
- Department of Pathology, University Hospital of Tübingen, 72076 Tübingen, Germany; and
| | - Jutta Gamper-Tsigaras
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tübingen, 72076 Tübingen, Germany
| | - Jörg Reutershan
- Department of Anesthesiology and Intensive Care Medicine, Bayreuth Hospital, 95445 Bayreuth, Germany
| | - Franziska M Konrad
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tübingen, 72076 Tübingen, Germany;
| |
Collapse
|
39
|
An mRNA expression signature for prognostication in de novo acute myeloid leukemia patients with normal karyotype. Oncotarget 2016; 6:39098-110. [PMID: 26517675 PMCID: PMC4770759 DOI: 10.18632/oncotarget.5390] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 08/30/2015] [Indexed: 12/28/2022] Open
Abstract
Although clinical features, cytogenetics, and mutations are widely used to predict prognosis in patients with acute myeloid leukemia (AML), further refinement of risk stratification is necessary for optimal treatment, especially in cytogenetically normal (CN) patients. We sought to generate a simple gene expression signature as a predictor of clinical outcome through analyzing the mRNA arrays of 158 de novo CN AML patients. We compared the gene expression profiles of patients with poor response to induction chemotherapy with those who responded well. Forty-six genes expressed differentially between the two groups. Among them, expression of 11 genes was significantly associated with overall survival (OS) in univariate Cox regression analysis in 104 patients who received standard intensive chemotherapy. We integrated the z-transformed expression levels of these 11 genes to generate a risk scoring system. Higher risk scores were significantly associated with shorter OS (median 17.0 months vs. not reached, P < 0.001) in ours and another 3 validation cohorts. In addition, it was an independent unfavorable prognostic factor by multivariate analysis (HR 1.116, 95% CI 1.035~1.204, P = 0.004). In conclusion, we developed a simple mRNA expression signature for prognostication in CN-AML patients. This prognostic biomarker will help refine the treatment strategies for this group of patients.
Collapse
|
40
|
Enciso J, Mayani H, Mendoza L, Pelayo R. Modeling the Pro-inflammatory Tumor Microenvironment in Acute Lymphoblastic Leukemia Predicts a Breakdown of Hematopoietic-Mesenchymal Communication Networks. Front Physiol 2016; 7:349. [PMID: 27594840 PMCID: PMC4990565 DOI: 10.3389/fphys.2016.00349] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 08/02/2016] [Indexed: 01/10/2023] Open
Abstract
Lineage fate decisions of hematopoietic cells depend on intrinsic factors and extrinsic signals provided by the bone marrow microenvironment, where they reside. Abnormalities in composition and function of hematopoietic niches have been proposed as key contributors of acute lymphoblastic leukemia (ALL) progression. Our previous experimental findings strongly suggest that pro-inflammatory cues contribute to mesenchymal niche abnormalities that result in maintenance of ALL precursor cells at the expense of normal hematopoiesis. Here, we propose a molecular regulatory network interconnecting the major communication pathways between hematopoietic stem and progenitor cells (HSPCs) and mesenchymal stromal cells (MSCs) within the BM. Dynamical analysis of the network as a Boolean model reveals two stationary states that can be interpreted as the intercellular contact status. Furthermore, simulations describe the molecular patterns observed during experimental proliferation and activation. Importantly, our model predicts instability in the CXCR4/CXCL12 and VLA4/VCAM1 interactions following microenvironmental perturbation due by temporal signaling from Toll like receptors (TLRs) ligation. Therefore, aberrant expression of NF-κB induced by intrinsic or extrinsic factors may contribute to create a tumor microenvironment where a negative feedback loop inhibiting CXCR4/CXCL12 and VLA4/VCAM1 cellular communication axes allows for the maintenance of malignant cells.
Collapse
Affiliation(s)
- Jennifer Enciso
- Oncology Research Unit, Mexican Institute for Social SecurityMexico City, Mexico; Biochemistry Sciences Program, Universidad Nacional Autónoma de MexicoMexico City, Mexico
| | - Hector Mayani
- Oncology Research Unit, Mexican Institute for Social Security Mexico City, Mexico
| | - Luis Mendoza
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de Mexico Mexico City, Mexico
| | - Rosana Pelayo
- Oncology Research Unit, Mexican Institute for Social Security Mexico City, Mexico
| |
Collapse
|
41
|
Behnam Azad B, Lisok A, Chatterjee S, Poirier JT, Pullambhatla M, Luker GD, Pomper MG, Nimmagadda S. Targeted Imaging of the Atypical Chemokine Receptor 3 (ACKR3/CXCR7) in Human Cancer Xenografts. J Nucl Med 2016; 57:981-8. [PMID: 26912435 PMCID: PMC5261856 DOI: 10.2967/jnumed.115.167932] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/08/2016] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The atypical chemokine receptor ACKR3 (formerly CXCR7), overexpressed in various cancers compared with normal tissues, plays a pivotal role in adhesion, angiogenesis, tumorigenesis, metastasis, and tumor cell survival. ACKR3 modulates the tumor microenvironment and regulates tumor growth. The therapeutic potential of ACKR3 has also been demonstrated in various murine models of human cancer. Literature findings underscore the importance of ACKR3 in disease progression and suggest it as an important diagnostic marker for noninvasive imaging of ACKR3-overexpressing malignancies. There are currently no reports on direct receptor-specific detection of ACKR3 expression. Here we report the evaluation of a radiolabeled ACKR3-targeted monoclonal antibody (ACKR3-mAb) for the noninvasive in vivo nuclear imaging of ACKR3 expression in human breast, lung, and esophageal squamous cell carcinoma cancer xenografts. METHODS ACKR3 expression data were extracted from Cancer Cell Line Encyclopedia, The Cancer Genome Atlas, and the Clinical Lung Cancer Genome Project. (89)Zr-ACKR3-mAb was evaluated in vitro and subsequently in vivo by PET and ex vivo biodistribution studies in mice xenografted with breast (MDA-MB-231-ACKR3 [231-ACKR3], MDA-MB-231 [231], MCF7), lung (HCC95), or esophageal (KYSE520) cancer cells. In addition, ACKR3-mAb was radiolabeled with (125)I and evaluated by SPECT imaging and ex vivo biodistribution studies. RESULTS ACKR3 transcript levels were highest in lung squamous cell carcinoma among the 21 cancer type data extracted from The Cancer Genome Atlas. Also, Clinical Lung Cancer Genome Project data showed that lung squamous cell carcinoma had the highest CXCR7 transcript levels compared with other lung cancer subtypes. The (89)Zr-ACKR3-mAb was produced in 80% ± 5% radiochemical yields with greater than 98% radiochemical purity. In vitro cell uptake of (89)Zr-ACKR3-mAb correlated with gradient levels of cell surface ACKR3 expression observed by flow cytometry. In vivo PET imaging and ex vivo biodistribution studies in mice with breast, lung, and esophageal cancer xenografts consistently showed enhanced (89)Zr-ACKR3-mAb uptake in high-ACKR3-expressing tumors. SPECT imaging of (125)I-ACKR3-mAb showed the versatility of ACKR3-mAb for in vivo monitoring of ACKR3 expression. CONCLUSION Data from this study suggest ACKR3 to be a viable diagnostic marker and demonstrate the utility of radiolabeled ACKR3-mAb for in vivo visualization of ACKR3-overexpressing malignancies.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/pharmacokinetics
- Biological Transport
- Cell Line, Tumor
- Cell Transformation, Neoplastic
- Female
- Humans
- Mice
- Molecular Imaging/methods
- Positron-Emission Tomography
- Radioisotopes
- Receptors, CXCR/immunology
- Receptors, CXCR/metabolism
- Tissue Distribution
- Tomography, Emission-Computed, Single-Photon
- Zirconium/chemistry
Collapse
Affiliation(s)
- Babak Behnam Azad
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Ala Lisok
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Samit Chatterjee
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - John T Poirier
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mrudula Pullambhatla
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland
| | - Gary D Luker
- Department of Radiology, University of Michigan, Ann Arbor, Michigan; and
| | - Martin G Pomper
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Sridhar Nimmagadda
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
42
|
Randhawa S, Cho BS, Ghosh D, Sivina M, Koehrer S, Müschen M, Peled A, Davis RE, Konopleva M, Burger JA. Effects of pharmacological and genetic disruption of CXCR4 chemokine receptor function in B-cell acute lymphoblastic leukaemia. Br J Haematol 2016; 174:425-36. [PMID: 27071778 DOI: 10.1111/bjh.14075] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/26/2016] [Indexed: 12/14/2022]
Abstract
B cell acute lymphoblastic leukaemia (B-ALL) cells express high levels of CXCR4 chemokine receptors for homing and retention within the marrow microenvironment. Bone marrow stromal cells (BMSC) secrete CXCL12, the ligand for CXCR4, and protect B-ALL cells from cytotoxic drugs. Therefore, the therapeutic use of CXCR4 antagonists has been proposed to disrupt cross talk between B-ALL cells and the protective stroma. Because CXCR4 antagonists can have activating agonistic function, we compared the genetic and pharmacological deletion of CXCR4 in B-ALL cells, using CRISPR-Cas9 gene editing and CXCR4 antagonists that are in clinical use (plerixafor, BKT140). Both genetic and pharmacological CXCR4 inhibition significantly reduced B-ALL cell migration to CXCL12 gradients and beneath BMSC, and restored drug sensitivity to dexamethasone, vincristine and cyclophosphamide. NOD/SCID/IL-2rγnull mice injected with CXCR4 gene-deleted B-ALL cells had significant delay in disease progression and superior survival when compared to control mice injected with CXCR4 wild-type B-ALL cells. These findings indicate that anti-leukaemia activity of CXCR4 antagonists is primarily due to CXCR4 inhibition, rather than agonistic activity, and corroborate that CXCR4 is an important target to overcome stroma-mediated drug resistance in B-ALL.
Collapse
Affiliation(s)
- Shubhchintan Randhawa
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Byung S Cho
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,Department of Haematology, Catholic Blood and Marrow Transplantation Centre, Cancer Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Dipanjan Ghosh
- Department of Lymphoma, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Mariela Sivina
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Stefan Koehrer
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Markus Müschen
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Amnon Peled
- Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Richard E Davis
- Department of Lymphoma, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jan A Burger
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
43
|
Vacchini A, Locati M, Borroni EM. Overview and potential unifying themes of the atypical chemokine receptor family. J Leukoc Biol 2016; 99:883-92. [PMID: 26740381 DOI: 10.1189/jlb.2mr1015-477r] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/12/2015] [Indexed: 12/17/2022] Open
Abstract
Chemokines modulate immune responses through their ability to orchestrate the migration of target cells. Chemokines directly induce cell migration through a distinct set of 7 transmembrane domain G protein-coupled receptors but are also recognized by a small subfamily of atypical chemokine receptors, characterized by their inability to support chemotactic activity. Atypical chemokine receptors are now emerging as crucial regulatory components of chemokine networks in a wide range of physiologic and pathologic contexts. Although a new nomenclature has been approved recently to reflect their functional distinction from their conventional counterparts, a systematic view of this subfamily is still missing. This review discusses their biochemical and immunologic properties to identify potential unifying themes in this emerging family.
Collapse
Affiliation(s)
- Alessandro Vacchini
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, and Humanitas Clinical and Research Center, Milan, Italy
| | - Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, and Humanitas Clinical and Research Center, Milan, Italy
| | - Elena Monica Borroni
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, and Humanitas Clinical and Research Center, Milan, Italy
| |
Collapse
|
44
|
Mikosik A, Henc I, Ruckemann-Dziurdzińska K, Frąckowiak JE, Płoszyńska A, Balcerska A, Bryl E, Witkowski JM. Increased μ-Calpain Activity in Blasts of Common B-Precursor Childhood Acute Lymphoblastic Leukemia Correlates with Their Lower Susceptibility to Apoptosis. PLoS One 2015; 10:e0136615. [PMID: 26317226 PMCID: PMC4552652 DOI: 10.1371/journal.pone.0136615] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 07/23/2015] [Indexed: 12/11/2022] Open
Abstract
Childhood acute lymphoblastic leukemia (ALL) blasts are characterized by inhibited apoptosis promoting fast disease progress. It is known that in chronic lymphocytic and acute myeloid leukemias the reduced apoptosis is strongly related with the activity of calpain-calpastatin system (CCS) composed of cytoplasmic proteases--calpains--performing the modulatory proteolysis of key proteins involved in cell proliferation and apoptosis, and of their endogenous inhibitor--calpastatin. Here, the CCS protein abundance and activity was for the first time studied in childhood ALL blasts and in control bone marrow CD19+ B cells by semi-quantitative flow cytometry and western blotting of calpastatin fragments resulting from endogenous calpain activity. Significantly higher μ-calpain (CAPN1) gene transcription, protein amounts and activity (but not those of m-calpain), with calpastatin amount and transcription of its gene (CAST) greatly varying were observed in CD19(+) ALL blasts compared to control cells. Significant inverse relation between the amount/activity of calpain and spontaneous apoptosis was noted. Patients older than 10 years (considered at higher risk) displayed increased amounts and activities of blast calpain. Finally, treatment of blasts with the tripeptide calpain inhibitors II and IV significantly and in dose-dependent fashion increased the percentage of blasts entering apoptosis. Together, these findings make the CCS a potential new predictive tool and therapeutic target in childhood ALL.
Collapse
Affiliation(s)
- Anna Mikosik
- Department of Pathophysiology, Medical University of Gdańsk, Gdańsk, Poland
| | - Izabella Henc
- Department of Pathology and Experimental Rheumatology, Medical University of Gdańsk, Gdańsk, Poland
| | | | | | - Anna Płoszyńska
- Clinic of Pediatrics, Hematology and Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | - Anna Balcerska
- Clinic of Pediatrics, Hematology and Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | - Ewa Bryl
- Department of Pathology and Experimental Rheumatology, Medical University of Gdańsk, Gdańsk, Poland
| | - Jacek M. Witkowski
- Department of Pathophysiology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
45
|
Alampour-Rajabi S, El Bounkari O, Rot A, Müller-Newen G, Bachelerie F, Gawaz M, Weber C, Schober A, Bernhagen J. MIF interacts with CXCR7 to promote receptor internalization, ERK1/2 and ZAP-70 signaling, and lymphocyte chemotaxis. FASEB J 2015; 29:4497-511. [PMID: 26139098 DOI: 10.1096/fj.15-273904] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 06/30/2015] [Indexed: 11/11/2022]
Abstract
Macrophage migration-inhibitory factor (MIF) is a pleiotropic cytokine with chemokine-like functions and is a mediator in numerous inflammatory conditions. Depending on the context, MIF signals through 1 or more of its receptors cluster of differentiation (CD)74, CXC-motif chemokine receptor (CXCR)2, and CXCR4. In addition, heteromeric receptor complexes have been identified. We characterized the atypical chemokine receptor CXCR7 as a novel receptor for MIF. MIF promoted human CXCR7 internalization up to 40%, peaking at 50-400 nM and 30 min, but CXCR7 internalization by MIF was not dependent on CXCR4. Yet, by coimmunoprecipitation, fluorescence microscopy, and a proximity ligation assay, CXCR7 was found to engage in MIF receptor complexes with CXCR4 and CD74, both after ectopic overexpression and in endogenous conditions in a human B-cell line. Receptor competition binding and coimmunoprecipitation studies combined with sulfo-SBED-biotin-transfer provided evidence for a direct interaction between MIF and CXCR7. Finally, we demonstrated MIF/CXCR7-mediated functional responses. Blockade of CXCR7 suppressed MIF-mediated ERK- and zeta-chain-associated protein kinase (ZAP)-70 activation (from 2.1- to 1.2-fold and from 2.5- to 1.6-fold, respectively) and fully abrogated primary murine B-cell chemotaxis triggered by MIF, but not by CXCL12. B cells from Cxcr7(-/-) mice exhibited an ablated transmigration response to MIF, indicating that CXCR7 is essential for MIF-promoted B-cell migration. Our findings provide biochemical and functional evidence that MIF is an alternative ligand of CXCR7 and suggest a functional role of the MIF-CXCR7 axis in B-lymphocyte migration.
Collapse
Affiliation(s)
- Setareh Alampour-Rajabi
- *Institute of Biochemistry and Molecular Cell Biology, Institute of Biochemistry and Molecular Biology, and Interdisziplinäres Zentrum für Klinische Forschung (IZKF), Rhine-Westphalia Technical University of Aachen (RWTH), Aachen, Germany; Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom; INSERM, Unité Mixte de Recherche-S 996, Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Université Paris-Sud, Clamart, France; Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, University of Tübingen, Tübingen, Germany; Institute for Cardiovascular Prevention, Klinikum der Universität München, and August-Lenz-Stiftung, Ludwig-Maximilians-Universität München, Munich, Germany; **Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; and Deutches Zentrum für Herz-Kreislauf Forschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Omar El Bounkari
- *Institute of Biochemistry and Molecular Cell Biology, Institute of Biochemistry and Molecular Biology, and Interdisziplinäres Zentrum für Klinische Forschung (IZKF), Rhine-Westphalia Technical University of Aachen (RWTH), Aachen, Germany; Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom; INSERM, Unité Mixte de Recherche-S 996, Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Université Paris-Sud, Clamart, France; Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, University of Tübingen, Tübingen, Germany; Institute for Cardiovascular Prevention, Klinikum der Universität München, and August-Lenz-Stiftung, Ludwig-Maximilians-Universität München, Munich, Germany; **Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; and Deutches Zentrum für Herz-Kreislauf Forschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Antal Rot
- *Institute of Biochemistry and Molecular Cell Biology, Institute of Biochemistry and Molecular Biology, and Interdisziplinäres Zentrum für Klinische Forschung (IZKF), Rhine-Westphalia Technical University of Aachen (RWTH), Aachen, Germany; Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom; INSERM, Unité Mixte de Recherche-S 996, Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Université Paris-Sud, Clamart, France; Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, University of Tübingen, Tübingen, Germany; Institute for Cardiovascular Prevention, Klinikum der Universität München, and August-Lenz-Stiftung, Ludwig-Maximilians-Universität München, Munich, Germany; **Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; and Deutches Zentrum für Herz-Kreislauf Forschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Gerhard Müller-Newen
- *Institute of Biochemistry and Molecular Cell Biology, Institute of Biochemistry and Molecular Biology, and Interdisziplinäres Zentrum für Klinische Forschung (IZKF), Rhine-Westphalia Technical University of Aachen (RWTH), Aachen, Germany; Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom; INSERM, Unité Mixte de Recherche-S 996, Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Université Paris-Sud, Clamart, France; Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, University of Tübingen, Tübingen, Germany; Institute for Cardiovascular Prevention, Klinikum der Universität München, and August-Lenz-Stiftung, Ludwig-Maximilians-Universität München, Munich, Germany; **Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; and Deutches Zentrum für Herz-Kreislauf Forschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Françoise Bachelerie
- *Institute of Biochemistry and Molecular Cell Biology, Institute of Biochemistry and Molecular Biology, and Interdisziplinäres Zentrum für Klinische Forschung (IZKF), Rhine-Westphalia Technical University of Aachen (RWTH), Aachen, Germany; Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom; INSERM, Unité Mixte de Recherche-S 996, Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Université Paris-Sud, Clamart, France; Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, University of Tübingen, Tübingen, Germany; Institute for Cardiovascular Prevention, Klinikum der Universität München, and August-Lenz-Stiftung, Ludwig-Maximilians-Universität München, Munich, Germany; **Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; and Deutches Zentrum für Herz-Kreislauf Forschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Meinrad Gawaz
- *Institute of Biochemistry and Molecular Cell Biology, Institute of Biochemistry and Molecular Biology, and Interdisziplinäres Zentrum für Klinische Forschung (IZKF), Rhine-Westphalia Technical University of Aachen (RWTH), Aachen, Germany; Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom; INSERM, Unité Mixte de Recherche-S 996, Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Université Paris-Sud, Clamart, France; Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, University of Tübingen, Tübingen, Germany; Institute for Cardiovascular Prevention, Klinikum der Universität München, and August-Lenz-Stiftung, Ludwig-Maximilians-Universität München, Munich, Germany; **Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; and Deutches Zentrum für Herz-Kreislauf Forschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christian Weber
- *Institute of Biochemistry and Molecular Cell Biology, Institute of Biochemistry and Molecular Biology, and Interdisziplinäres Zentrum für Klinische Forschung (IZKF), Rhine-Westphalia Technical University of Aachen (RWTH), Aachen, Germany; Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom; INSERM, Unité Mixte de Recherche-S 996, Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Université Paris-Sud, Clamart, France; Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, University of Tübingen, Tübingen, Germany; Institute for Cardiovascular Prevention, Klinikum der Universität München, and August-Lenz-Stiftung, Ludwig-Maximilians-Universität München, Munich, Germany; **Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; and Deutches Zentrum für Herz-Kreislauf Forschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Andreas Schober
- *Institute of Biochemistry and Molecular Cell Biology, Institute of Biochemistry and Molecular Biology, and Interdisziplinäres Zentrum für Klinische Forschung (IZKF), Rhine-Westphalia Technical University of Aachen (RWTH), Aachen, Germany; Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom; INSERM, Unité Mixte de Recherche-S 996, Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Université Paris-Sud, Clamart, France; Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, University of Tübingen, Tübingen, Germany; Institute for Cardiovascular Prevention, Klinikum der Universität München, and August-Lenz-Stiftung, Ludwig-Maximilians-Universität München, Munich, Germany; **Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; and Deutches Zentrum für Herz-Kreislauf Forschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Jürgen Bernhagen
- *Institute of Biochemistry and Molecular Cell Biology, Institute of Biochemistry and Molecular Biology, and Interdisziplinäres Zentrum für Klinische Forschung (IZKF), Rhine-Westphalia Technical University of Aachen (RWTH), Aachen, Germany; Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom; INSERM, Unité Mixte de Recherche-S 996, Laboratory of Excellence in Research on Medication and Innovative Therapeutics, Université Paris-Sud, Clamart, France; Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, University of Tübingen, Tübingen, Germany; Institute for Cardiovascular Prevention, Klinikum der Universität München, and August-Lenz-Stiftung, Ludwig-Maximilians-Universität München, Munich, Germany; **Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; and Deutches Zentrum für Herz-Kreislauf Forschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
46
|
Coste C, Neirinckx V, Gothot A, Wislet S, Rogister B. Are neural crest stem cells the missing link between hematopoietic and neurogenic niches? Front Cell Neurosci 2015; 9:218. [PMID: 26136659 PMCID: PMC4469833 DOI: 10.3389/fncel.2015.00218] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/22/2015] [Indexed: 12/24/2022] Open
Abstract
Hematopoietic niches are defined as cellular and molecular microenvironments that regulate hematopoietic stem cell (HSC) function together with stem cell autonomous mechanisms. Many different cell types have been characterized as contributors to the formation of HSC niches, such as osteoblasts, endothelial cells, Schwann cells, and mesenchymal progenitors. These mesenchymal progenitors have themselves been classified as CXC chemokine ligand (CXCL) 12-abundant reticular (CAR) cells, stem cell factor expressing cells, or nestin-positive mesenchymal stem cells (MSCs), which have been recently identified as neural crest-derived cells (NCSCs). Together, these cells are spatially associated with HSCs and believed to provide appropriate microenvironments for HSC self-renewal, differentiation, mobilization and hibernation both by cell-cell contact and soluble factors. Interestingly, it appears that regulatory pathways governing the hematopoietic niche homeostasis are operating in the neurogenic niche as well. Therefore, this review paper aims to compare both the regulation of hematopoietic and neurogenic niches, in order to highlight the role of NCSCs and nervous system components in the development and the regulation of the hematopoietic system.
Collapse
Affiliation(s)
- Cécile Coste
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - Virginie Neirinckx
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - André Gothot
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Cardiovascular Sciences, University of Liège Liège, Belgium ; Hematology Department, University Hospital Liège, Belgium
| | - Sabine Wislet
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - Bernard Rogister
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium ; Groupe Interdisciplinaire de Génoprotéomique Appliquée-Development, Stem Cells and Regenerative Medicine, University of Liège Liège, Belgium ; Neurology Department, University Hospital Liège, Belgium
| |
Collapse
|
47
|
Liesveld J. Plerixafor: potential role in acute leukemia therapy. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1020297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|