1
|
Safi R, Mohsen-Kanson T, Kouzi F, El-Saghir J, Dermesrobian V, Zugasti I, Zibara K, Menéndez P, El Hajj H, El-Sabban M. Direct Interaction Between CD34 + Hematopoietic Stem Cells and Mesenchymal Stem Cells Reciprocally Preserves Stemness. Cancers (Basel) 2024; 16:3972. [PMID: 39682159 DOI: 10.3390/cancers16233972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES A specialized microenvironment in the bone marrow, composed of stromal cells including mesenchymal stem cells (MSCs), supports hematopoietic stem cell (HSC) self-renewal, and differentiation bands play an important role in leukemia development and progression. The reciprocal direct interaction between MSCs and CD34+ HSCs under physiological and pathological conditions is yet to be fully characterized. METHODS Here, we established a direct co-culture model between MSCs and CD34+ HSCs or MSCs and acute myeloid leukemia cells (THP-1, Molm-13, and primary cells from patients) to study heterocellular communication. RESULTS Following MSCs-CD34+ HSCs co-culture, the expression of adhesion markers N-Cadherin and connexin 43 increased in both cell types, forming gap junction channels. Moreover, the clonogenic potential of CD34+ HSCs was increased. However, direct contact of acute myeloid leukemia cells with MSCs reduced the expression levels of connexin 43 and N-Cadherin in MSCs. The impairment in gap junction formation may potentially be due to a defect in the acute myeloid leukemia-derived MSCs. Interestingly, CD34+ HSCs and acute myeloid leukemia cell lines attenuated MSC osteoblastic differentiation upon prolonged direct cell-cell contact. CONCLUSIONS In conclusion, under physiological conditions, connexin 43 and N-Cadherin interaction preserves stemness of both CD34+ HSCs and MSCs, a process that is compromised in acute myeloid leukemia, pointing to the possible role of gap junctions in modulating stemness.
Collapse
Affiliation(s)
- Rémi Safi
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
- Josep Carreras Leukemia Research Institute, 08916 Barcelona, Spain
| | - Tala Mohsen-Kanson
- Faculty of Science, Lebanese University, Zahle 1801, Lebanon
- Faculty of Science, Lebanese University, Hadath 40016, Lebanon
| | - Farah Kouzi
- Faculty of Science, Lebanese University, Zahle 1801, Lebanon
- Faculty of Science, Lebanese University, Hadath 40016, Lebanon
| | - Jamal El-Saghir
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vera Dermesrobian
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
- Laboratory of Adaptive Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Inés Zugasti
- Department of Hematology, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| | - Kazem Zibara
- Faculty of Science, Lebanese University, Zahle 1801, Lebanon
- Faculty of Science, Lebanese University, Hadath 40016, Lebanon
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, 08916 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
- Consorcio Investigación Biomédica en Red de Cancer, CIBER-ONC, ISCIII, 28029 Barcelona, Spain
- Spanish Network for Advanced Cell Therapies (TERAV), 08028 Barcelona, Spain
| | - Hiba El Hajj
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
| |
Collapse
|
2
|
Busch C, Mulholland T, Zagnoni M, Dalby M, Berry C, Wheadon H. Overcoming BCR::ABL1 dependent and independent survival mechanisms in chronic myeloid leukaemia using a multi-kinase targeting approach. Cell Commun Signal 2023; 21:342. [PMID: 38031192 PMCID: PMC10685629 DOI: 10.1186/s12964-023-01363-2] [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: 07/04/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Despite improved patient outcome using tyrosine kinase inhibitors (TKIs), chronic myeloid leukaemia (CML) patients require life-long treatment due to leukaemic stem cell (LSC) persistence. LSCs reside in the bone marrow (BM) niche, which they modify to their advantage. The BM provides oncogene-independent signals to aid LSC cell survival and quiescence. The bone-morphogenetic pathway (BMP) is one pathway identified to be highly deregulated in CML, with high levels of BMP ligands detected in the BM, accompanied by CML stem and progenitor cells overexpressing BMP type 1 receptors- activin-like kinases (ALKs), especially in TKI resistant patients. Saracatinib (SC), a SRC/ABL1 dual inhibitor, inhibits the growth of CML cells resistant to the TKI imatinib (IM). Recent studies indicate that SC is also a potent ALK inhibitor and BMP antagonist. Here we investigate the efficacy of SC in overcoming CML BCR::ABL1 dependent and independent signals mediated by the BM niche both in 2D and 3D culture. METHODS CML cells (K562 cell line and CML CD34+ primary cells) were treated with single or combination treatments of: IM, SC and the BMP receptors inhibitor dorsomorphin (DOR), with or without BMP4 stimulation in 2D (suspension) and 3D co-culture on HS5 stroma cell line and mesenchymal stem cells in AggreWell and microfluidic devices. Flow cytometry was performed to investigate apoptosis, cell cycle progression and proliferation, alongside colony assays following treatment. Proteins changes were validated by immunoblotting and transcriptional changes by Fluidigm multiplex qPCR. RESULTS By targeting the BMP pathway, using specific inhibitors against ALKs in combination with SRC and ABL TKIs, we show an increase in apoptosis, altered cell cycle regulation, fewer cell divisions, and reduced numbers of CD34+ cells. Impairment of long-term proliferation and differentiation potential after combinatorial treatment also occurred. CONCLUSION BMP signalling pathway is important for CML cell survival. Targeting SRC, ABL and ALK kinases is more effective than ABL inhibition alone, the combination efficacy importantly being demonstrated in both 2D and 3D cell cultures highlighting the need for combinatorial therapies in contrast to standard of care single agents. Our study provides justification to target multiple kinases in CML to combat LSC persistence.
Collapse
Affiliation(s)
- Caroline Busch
- Paul O'Gorman Leukaemia Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Theresa Mulholland
- Centre for Microsystems and Photonics, Electronic and Electrical Engineering, University of Strathclyde, Glasgow, G1 1XW, UK
| | - Michele Zagnoni
- Centre for Microsystems and Photonics, Electronic and Electrical Engineering, University of Strathclyde, Glasgow, G1 1XW, UK
| | - Matthew Dalby
- Mazumdar-Shaw Advanced Research Centre, School of Molecular Biosciences, University of Glasgow, Glasgow, G11 6EW, UK
| | - Catherine Berry
- Mazumdar-Shaw Advanced Research Centre, School of Molecular Biosciences, University of Glasgow, Glasgow, G11 6EW, UK
| | - Helen Wheadon
- Paul O'Gorman Leukaemia Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK.
| |
Collapse
|
3
|
Nevárez-Ramírez AJ, Guzmán-Ortiz AL, Cortes-Reynosa P, Perez-Salazar E, Jaimes-Ortega GA, Valle-Rios R, Marín-Hernández Á, Rodríguez-Zavala JS, Ruiz-May E, Castrejón-Flores JL, Quezada H. Shotgun Proteomics of Co-Cultured Leukemic and Bone Marrow Stromal Cells from Different Species as a Preliminary Approach to Detect Intercellular Protein Transfer. Proteomes 2023; 11:proteomes11020015. [PMID: 37092456 PMCID: PMC10123657 DOI: 10.3390/proteomes11020015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/25/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
Cellular interactions within the bone marrow microenvironment modulate the properties of subsets of leukemic cells leading to the development of drug-resistant phenotypes. The intercellular transfer of proteins and organelles contributes to this process but the set of transferred proteins and their effects in the receiving cells remain unclear. This study aimed to detect the intercellular protein transfer from mouse bone marrow stromal cells (OP9 cell line) to human T-lymphoblasts (CCRF-CEM cell line) using nanoLC-MS/MS-based shotgun proteomics in a 3D co-culture system. After 24 h of co-culture, 1513 and 67 proteins from human and mouse origin, respectively, were identified in CCRF-CEM cells. The presence of mouse proteins in the human cell line, detected by analyzing the differences in amino acid sequences of orthologous peptides, was interpreted as the result of intercellular transfer. The transferred proteins might have contributed to the observed resistance to vincristine, methotrexate, and hydrogen peroxide in the co-cultured leukemic cells. Our results suggest that shotgun proteomic analyses of co-cultured cells from different species could be a simple option to get a preliminary survey of the proteins exchanged among interacting cells.
Collapse
Affiliation(s)
- Abraham Josué Nevárez-Ramírez
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Dr. Márquez 162, Doctores, Mexico City 06720, Mexico
- Unidad Profesional Interdisciplinaria de Biotecnología, Instituto Politécnico Nacional, Av. Acueducto s/n, Barrio La Laguna, Mexico City 07340, Mexico
| | - Ana Laura Guzmán-Ortiz
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Dr. Márquez 162, Doctores, Mexico City 06720, Mexico
| | - Pedro Cortes-Reynosa
- Departamento de Biología Celular, CINVESTAV-IPN, Av Instituto Politécnico Nacional 2508, San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Eduardo Perez-Salazar
- Departamento de Biología Celular, CINVESTAV-IPN, Av Instituto Politécnico Nacional 2508, San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Gustavo Alberto Jaimes-Ortega
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Dr. Márquez 162, Doctores, Mexico City 06720, Mexico
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Circuito interior, Av. Universidad 3000, Ciudad Universitaria, Coyoacán, Mexico City 04510, Mexico
| | - Ricardo Valle-Rios
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Dr. Márquez 162, Doctores, Mexico City 06720, Mexico
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Circuito interior, Av. Universidad 3000, Ciudad Universitaria, Coyoacán, Mexico City 04510, Mexico
| | - Álvaro Marín-Hernández
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Belisario Domínguez—Sección XVI, Mexico City 14080, Mexico
| | - José S. Rodríguez-Zavala
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Belisario Domínguez—Sección XVI, Mexico City 14080, Mexico
| | - Eliel Ruiz-May
- Red de Estudios Moleculares Avanzados, Clúster Científico y Tecnológico BioMimic®, Instituto de Ecología A.C. (INECOL), Carretera Antigua a Coatepec 351, El Haya, Xalapa 91073, Mexico
| | - José Luis Castrejón-Flores
- Unidad Profesional Interdisciplinaria de Biotecnología, Instituto Politécnico Nacional, Av. Acueducto s/n, Barrio La Laguna, Mexico City 07340, Mexico
| | - Héctor Quezada
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Dr. Márquez 162, Doctores, Mexico City 06720, Mexico
| |
Collapse
|
4
|
Abubakar M, Fan S, Bowles EA, Widemann L, Duggan MA, Pfeiffer RM, Falk RT, Lawrence S, Richert-Boe K, Glass AG, Kimes TM, Figueroa JD, Rohan TE, Gierach GL. Relation of Quantitative Histologic and Radiologic Breast Tissue Composition Metrics With Invasive Breast Cancer Risk. JNCI Cancer Spectr 2021; 5:pkab015. [PMID: 33981950 PMCID: PMC8103888 DOI: 10.1093/jncics/pkab015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/09/2020] [Accepted: 02/01/2021] [Indexed: 12/18/2022] Open
Abstract
Background Benign breast disease (BBD) is a strong breast cancer risk factor, but identifying patients that might develop invasive breast cancer remains a challenge. Methods By applying machine-learning to digitized hematoxylin and eosin-stained biopsies and computer-assisted thresholding to mammograms obtained circa BBD diagnosis, we generated quantitative tissue composition metrics and determined their association with future invasive breast cancer diagnosis. Archival breast biopsies and mammograms were obtained for women (18-86 years of age) in a case-control study, nested within a cohort of 15 395 BBD patients from Kaiser Permanente Northwest (1970-2012), followed through mid-2015. Patients who developed incident invasive breast cancer (ie, cases; n = 514) and those who did not (ie, controls; n = 514) were matched on BBD diagnosis age and plan membership duration. All statistical tests were 2-sided. Results Increasing epithelial area on the BBD biopsy was associated with increasing breast cancer risk (odds ratio [OR]Q4 vs Q1 = 1.85, 95% confidence interval [CI] = 1.13 to 3.04; P trend = .02). Conversely, increasing stroma was associated with decreased risk in nonproliferative, but not proliferative, BBD (P heterogeneity = .002). Increasing epithelium-to-stroma proportion (ORQ4 vs Q1 = 2.06, 95% CI =1.28 to 3.33; P trend = .002) and percent mammographic density (MBD) (ORQ4 vs Q1 = 2.20, 95% CI = 1.20 to 4.03; P trend = .01) were independently and strongly predictive of increased breast cancer risk. In combination, women with high epithelium-to-stroma proportion and high MBD had substantially higher risk than those with low epithelium-to-stroma proportion and low MBD (OR = 2.27, 95% CI = 1.27 to 4.06; P trend = .005), particularly among women with nonproliferative (P trend = .01) vs proliferative (P trend = .33) BBD. Conclusion Among BBD patients, increasing epithelium-to-stroma proportion on BBD biopsies and percent MBD at BBD diagnosis were independently and jointly associated with increasing breast cancer risk. These findings were particularly striking for women with nonproliferative disease (comprising approximately 70% of all BBD patients), for whom relevant predictive biomarkers are lacking.
Collapse
Affiliation(s)
- Mustapha Abubakar
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, USA
- Correspondence to: Mustapha Abubakar, MD, PhD, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, 9609 Medical Center Drive, Rockville, MD, USA (e-mail: )
| | - Shaoqi Fan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, USA
| | - Erin Aiello Bowles
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Lea Widemann
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, USA
| | - Máire A Duggan
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, USA
| | - Roni T Falk
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, USA
| | - Scott Lawrence
- Molecular and Digital Pathology Laboratory, Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc, Frederick, MD, USA
| | | | - Andrew G Glass
- Kaiser Permanente Center for Health Research, Portland, OR, USA
| | - Teresa M Kimes
- Kaiser Permanente Center for Health Research, Portland, OR, USA
| | - Jonine D Figueroa
- Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Gretchen L Gierach
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, USA
| |
Collapse
|
5
|
Involvement of GPx-3 in the Reciprocal Control of Redox Metabolism in the Leukemic Niche. Int J Mol Sci 2020; 21:ijms21228584. [PMID: 33202543 PMCID: PMC7696155 DOI: 10.3390/ijms21228584] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 12/21/2022] Open
Abstract
The bone marrow (BM) microenvironment plays a crucial role in the development and progression of leukemia (AML). Intracellular reactive oxygen species (ROS) are involved in the regulation of the biology of leukemia-initiating cells, where the antioxidant enzyme GPx-3 could be involved as a determinant of cellular self-renewal. Little is known however about the role of the microenvironment in the control of the oxidative metabolism of AML cells. In the present study, a coculture model of BM mesenchymal stromal cells (MSCs) and AML cells (KG1a cell-line and primary BM blasts) was used to explore this metabolic pathway. MSC-contact, rather than culture with MSC-conditioned medium, decreases ROS levels and inhibits the Nrf-2 pathway through overexpression of GPx3 in AML cells. The decrease of ROS levels also inactivates p38MAPK and reduces the proliferation of AML cells. Conversely, contact with AML cells modifies MSCs in that they display an increased oxidative stress and Nrf-2 activation, together with a concomitant lowered expression of GPx-3. Altogether, these experiments suggest that a reciprocal control of oxidative metabolism is initiated by direct cell–cell contact between MSCs and AML cells. GPx-3 expression appears to play a crucial role in this cross-talk and could be involved in the regulation of leukemogenesis.
Collapse
|
6
|
Oxidative resistance of leukemic stem cells and oxidative damage to hematopoietic stem cells under pro-oxidative therapy. Cell Death Dis 2020; 11:291. [PMID: 32341354 PMCID: PMC7184730 DOI: 10.1038/s41419-020-2488-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/08/2020] [Accepted: 04/08/2020] [Indexed: 02/07/2023]
Abstract
Leukemic stem cells (LSCs) and hematopoietic stem cells (HSCs) are both dependent on the hypoxic bone marrow (BM) microenvironment (also known as the BM niche). There is always fierce competition between the two types of cells, and the former exhibits a greater competitive advantage than the latter via multiple mechanisms. Under hypoxia, the dynamic balance between the generation and clearing of intracellular reactive oxygen species (ROS) is conducive to maintaining a quiescent state of cells. Quiescent LSCs can reside well in the BM niche, avoiding attack by chemotherapeutic agents, which is the cause of chemotherapeutic resistance and relapse in leukemia. HSCs acquire energy mainly through anaerobic glycolysis, whereas LSCs achieve energy metabolism largely through mitochondrial oxidative respiration. Mitochondria are the primary site of ROS generation. Thus, in theory, mitochondria-mediated respiration will cause an increase in ROS generation in LSCs and a higher intracellular oxidative stress level. The sensitivity of the cells to pro-oxidant drugs increases as well, which allows for the selective clearing of LSCs by pro-oxidative therapy. However, HSCs are also highly sensitive to changes in ROS levels, and the toxic effects of pro-oxidant drugs on HSCs poses a major challenge to pro-oxidative therapy in leukemia. Given the above facts, we reviewed studies on the oxidative resistance of LSCs and the oxidative damage to HSCs under pro-oxidative therapy. An in-depth investigation into the oxidative stress status and regulatory mechanisms of LSCs and HSCs in hypoxic environments will promote our understanding of the survival strategy employed by LSCs and the mechanism of the oxidative damage to HSCs in the BM niche, thus facilitating individualized treatment of leukemia patients and helping eliminate LSCs without disturbing normal hematopoietic cells.
Collapse
|
7
|
Singh AK, Cancelas JA. Gap Junctions in the Bone Marrow Lympho-Hematopoietic Stem Cell Niche, Leukemia Progression, and Chemoresistance. Int J Mol Sci 2020; 21:E796. [PMID: 31991829 PMCID: PMC7038046 DOI: 10.3390/ijms21030796] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/19/2020] [Accepted: 01/23/2020] [Indexed: 12/15/2022] Open
Abstract
Abstract: The crosstalk between hematopoietic stem cells (HSC) and bone marrow (BM) microenvironment is critical for homeostasis and hematopoietic regeneration in response to blood formation emergencies after injury, and has been associated with leukemia transformation and progression. Intercellular signals by the BM stromal cells in the form of cell-bound or secreted factors, or by physical interaction, regulate HSC localization, maintenance, and differentiation within increasingly defined BM HSC niches. Gap junctions (GJ) are comprised of arrays of membrane embedded channels formed by connexin proteins, and control crucial signaling functions, including the transfer of ions, small metabolites, and organelles to adjacent cells which affect intracellular mechanisms of signaling and autophagy. This review will discuss the role of GJ in both normal and leukemic hematopoiesis, and highlight some of the most novel approaches that may improve the efficacy of cytotoxic drugs. Connexin GJ channels exert both cell-intrinsic and cell-extrinsic effects on HSC and BM stromal cells, involved in regenerative hematopoiesis after myelosuppression, and represent an alternative system of cell communication through a combination of electrical and metabolic coupling as well as organelle transfer in the HSC niche. GJ intercellular communication (GJIC) in the HSC niche improves cellular bioenergetics, and rejuvenates damaged recipient cells. Unfortunately, they can also support leukemia proliferation and survival by creating leukemic niches that provide GJIC dependent energy sources and facilitate chemoresistance and relapse. The emergence of new strategies to disrupt self-reinforcing malignant niches and intercellular organelle exchange in leukemic niches, while at the same time conserving normal hematopoietic GJIC function, could synergize the effect of chemotherapy drugs in eradicating minimal residual disease. An improved understanding of the molecular basis of connexin regulation in normal and leukemic hematopoiesis is warranted for the re-establishment of normal hematopoiesis after chemotherapy.
Collapse
Affiliation(s)
- Abhishek K. Singh
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229, USA;
- Hoxworth Blood Center, University of Cincinnati Academic Health Center, 3333 Burnet Ave., Cincinnati, OH 45229, USA
| | - Jose A. Cancelas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229, USA;
- Hoxworth Blood Center, University of Cincinnati Academic Health Center, 3333 Burnet Ave., Cincinnati, OH 45229, USA
| |
Collapse
|
8
|
Kolba MD, Dudka W, Zaręba-Kozioł M, Kominek A, Ronchi P, Turos L, Chroscicki P, Wlodarczyk J, Schwab Y, Klejman A, Cysewski D, Srpan K, Davis DM, Piwocka K. Tunneling nanotube-mediated intercellular vesicle and protein transfer in the stroma-provided imatinib resistance in chronic myeloid leukemia cells. Cell Death Dis 2019; 10:817. [PMID: 31659149 PMCID: PMC6817823 DOI: 10.1038/s41419-019-2045-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 09/25/2019] [Accepted: 10/08/2019] [Indexed: 01/16/2023]
Abstract
Intercellular communication within the bone marrow niche significantly promotes leukemogenesis and provides protection of leukemic cells from therapy. Secreted factors, intercellular transfer of mitochondria and the receptor-ligand interactions have been shown as mediators of this protection. Here we report that tunneling nanotubes (TNTs)-long, thin membranous structures, which have been identified as a novel mode of intercellular cross-talk-are formed in the presence of stroma and mediate transfer of cellular vesicles from stroma to leukemic cells. Importantly, transmission of vesicles via TNTs from stromal cells increases resistance of leukemic cells to the tyrosine kinase inhibitor, imatinib. Using correlative light-electron microscopy and electron tomography we show that stromal TNTs contain vesicles, provide membrane continuity with the cell bodies and can be open-ended. Moreover, trans-SILAC studies to reveal the non-autonomous proteome showed that specific sets of proteins are transferred together with cellular vesicles from stromal to leukemic cells, with a potential role in survival and adaptation. Altogether, our findings provide evidence for the biological role of the TNT-mediated vesicle exchange between stromal and leukemic cells, implicating the direct vesicle and protein transfer in the stroma-provided protection of leukemic cells.
Collapse
Affiliation(s)
- Marta D Kolba
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Wioleta Dudka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Monika Zaręba-Kozioł
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Agata Kominek
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Paolo Ronchi
- Electron Microscopy Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Laura Turos
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Piotr Chroscicki
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Jakub Wlodarczyk
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Yannick Schwab
- Electron Microscopy Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany.,Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Agata Klejman
- Laboratory of Animal Models, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Dominik Cysewski
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Katja Srpan
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| | - Daniel M Davis
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
9
|
Kouzi F, Zibara K, Bourgeais J, Picou F, Gallay N, Brossaud J, Dakik H, Roux B, Hamard S, Le Nail LR, Hleihel R, Foucault A, Ravalet N, Rouleux-Bonnin F, Gouilleux F, Mazurier F, Bene MC, Akl H, Gyan E, Domenech J, El-Sabban M, Herault O. Disruption of gap junctions attenuates acute myeloid leukemia chemoresistance induced by bone marrow mesenchymal stromal cells. Oncogene 2019; 39:1198-1212. [PMID: 31649334 PMCID: PMC7002301 DOI: 10.1038/s41388-019-1069-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 01/09/2023]
Abstract
The bone marrow (BM) niche impacts the progression of acute myeloid leukemia (AML) by favoring the chemoresistance of AML cells. Intimate interactions between leukemic cells and BM mesenchymal stromal cells (BM-MSCs) play key roles in this process. Direct intercellular communications between hematopoietic cells and BM-MSCs involve connexins, components of gap junctions. We postulated that blocking gap junction assembly could modify cell–cell interactions in the leukemic niche and consequently the chemoresistance. The comparison of BM-MSCs from AML patients and healthy donors revealed a specific profile of connexins in BM-MSCs of the leukemic niche and the effects of carbenoxolone (CBX), a gap junction disruptor, were evaluated on AML cells. CBX presents an antileukemic effect without affecting normal BM-CD34+ progenitor cells. The proapoptotic effect of CBX on AML cells is in line with the extinction of energy metabolism. CBX acts synergistically with cytarabine (Ara-C) in vitro and in vivo. Coculture experiments of AML cells with BM-MSCs revealed that CBX neutralizes the protective effect of the niche against the Ara-C-induced apoptosis of leukemic cells. Altogether, these results suggest that CBX could be of therapeutic interest to reduce the chemoresistance favored by the leukemic niche, by targeting gap junctions, without affecting normal hematopoiesis.
Collapse
Affiliation(s)
- Farah Kouzi
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,PRASE, DSST, Lebanese University, Beirut, Lebanon
| | - Kazem Zibara
- PRASE, DSST, Lebanese University, Beirut, Lebanon.,Biology Department, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Jerome Bourgeais
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| | - Frederic Picou
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| | - Nathalie Gallay
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| | - Julie Brossaud
- Department of Nuclear Medicine, Bordeaux University Hospital, Pessac, France
| | - Hassan Dakik
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France
| | - Benjamin Roux
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| | - Sophie Hamard
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France
| | | | - Rita Hleihel
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Amelie Foucault
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| | - Noemie Ravalet
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| | - Florence Rouleux-Bonnin
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France
| | - Fabrice Gouilleux
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France
| | - Frederic Mazurier
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France
| | - Marie C Bene
- Department of Biological Hematology, Nantes University Hospital, CRCINA, Nantes, France
| | - Haidar Akl
- PRASE, DSST, Lebanese University, Beirut, Lebanon.,Biology Department, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Emmanuel Gyan
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Hematology and Cell Therapy, Tours University Hospital, Tours, France
| | - Jorge Domenech
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Olivier Herault
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France. .,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France. .,Department of Biological Hematology, Tours University Hospital, Tours, France.
| |
Collapse
|
10
|
Gaudichon J, Jakobczyk H, Debaize L, Cousin E, Galibert MD, Troadec MB, Gandemer V. Mechanisms of extramedullary relapse in acute lymphoblastic leukemia: Reconciling biological concepts and clinical issues. Blood Rev 2019; 36:40-56. [PMID: 31010660 DOI: 10.1016/j.blre.2019.04.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 04/03/2019] [Accepted: 04/15/2019] [Indexed: 12/17/2022]
Abstract
Long-term survival rates in childhood acute lymphoblastic leukemia (ALL) are currently above 85% due to huge improvements in treatment. However, 15-20% of children still experience relapses. Relapses can either occur in the bone marrow or at extramedullary sites, such as gonads or the central nervous system (CNS), formerly referred to as ALL-blast sanctuaries. The reason why ALL cells migrate to and stay in these sites is still unclear. In this review, we have attempted to assemble the evidence concerning the microenvironmental factors that could explain why ALL cells reside in such sites. We present criteria that make extramedullary leukemia niches and solid tumor metastatic niches comparable. Indeed, considering extramedullary leukemias as metastases could be a useful approach for proposing more effective treatments. In this context, we conclude with several examples of potential niche-based therapies which could be successfully added to current treatments of ALL.
Collapse
Affiliation(s)
- Jérémie Gaudichon
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France; Pediatric Hematology and Oncology Department, University Hospital, Caen, France.
| | - Hélène Jakobczyk
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France
| | - Lydie Debaize
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France
| | - Elie Cousin
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France; Pediatric Hematology Department, University Hospital, Rennes, France
| | - Marie-Dominique Galibert
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France.
| | - Marie-Bérengère Troadec
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France
| | - Virginie Gandemer
- CNRS, IGDR (Institut de Génétique et Développement de Rennes), Univ Rennes, UMR 6290, Rennes F-35000, France; Pediatric Hematology Department, University Hospital, Rennes, France.
| |
Collapse
|
11
|
Ding L, Zhang W, Yang L, Pelicano H, Zhou K, Yin R, Huang R, Zeng J. Targeting the autophagy in bone marrow stromal cells overcomes resistance to vorinostat in chronic lymphocytic leukemia. Onco Targets Ther 2018; 11:5151-5170. [PMID: 30210236 PMCID: PMC6114474 DOI: 10.2147/ott.s170392] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background The bone marrow microenvironment constitutes a sanctuary for leukemia cells. Recent evidence indicates that environment-mediated drug resistance arises from a reciprocal influence between tumor cells and the surrounding stroma. The present study aimed to investigate the effect of chronic lymphocytic leukemia (CLL) cells on the metabolism of bone marrow stroma, to determine the role of this metabolic change in the stroma in vorinostat resistance of CLL cells, and thus to assess a novel strategy to target stroma and achieve the maximum therapeutic effect of vorinostat. Methods To evaluate this issue, we used freshly isolated CLL cells from peripheral blood samples of patients with CLL, and co-cultured them with bone marrow stromal cell lines to examine autophagy activity and metabolic changes in both CLL cells and stromal cells after vorinostat treatment. Results The results demonstrated that CLL cells were under intrinsic oxidative stress which was further enhanced by vorinostat treatment, and released H2O2 outside the cells. The adjacent stromal cells took up H2O2 and drove autophagy, mitophagy and glycolysis, resulting in the local production of high-energy mitochondrial fuels, which were then taken up by CLL cells to be effectively utilized through mitochondrial oxidative phosphorylation to enable more ATP production. Notably, targeting autophagic stromal cells with autophagy inhibitor remarkably decreased stromal protection against vorinostat treatment in CLL cells. Conclusion This study demonstrated that the stroma in the CLL microenvironment is abnormal and undergoes autophagy, and manipulation of autophagic stromal cells could serve as a novel promising strategy to circumvent stroma-mediated drug resistance in CLL cells.
Collapse
Affiliation(s)
- Lu Ding
- Department of Leukemia, The First Affiliated Hospital of Nanchang University, Nanchang, China, .,School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Wan Zhang
- Department of Leukemia, The First Affiliated Hospital of Nanchang University, Nanchang, China,
| | - Lili Yang
- Department of Hematology, People's Hospital of Jiangxi Province, Nanchang, China
| | - Helene Pelicano
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,
| | - Kaiwen Zhou
- The First Clinical Medical College, School of Medicine, Nanchang University, Nanchang, China
| | - Ran Yin
- Department of Leukemia, The First Affiliated Hospital of Nanchang University, Nanchang, China,
| | - Ruibin Huang
- Department of Leukemia, The First Affiliated Hospital of Nanchang University, Nanchang, China,
| | - Junyi Zeng
- Department of Leukemia, The First Affiliated Hospital of Nanchang University, Nanchang, China,
| |
Collapse
|
12
|
Gleisner MA, Navarrete M, Hofmann F, Salazar-Onfray F, Tittarelli A. Mind the Gaps in Tumor Immunity: Impact of Connexin-Mediated Intercellular Connections. Front Immunol 2017; 8:1067. [PMID: 28919895 PMCID: PMC5585150 DOI: 10.3389/fimmu.2017.01067] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 08/16/2017] [Indexed: 12/22/2022] Open
Abstract
Gap junctions (GJs)-mediated intercellular communications (GJICs) are connexin (Cx)-formed plasma membrane channels that allow for the passage of small molecules between adjacent cells, and are involved in several physiopathological processes, including immune responses against cancer. In general, tumor cells are poorly coupled through GJs, mainly due to low Cx expression or reduced channel activity, suggesting that Cxs may have tumor suppressor roles. However, more recent data indicate that Cxs and/or GJICs may also in some cases promote tumor progression. This dual role of Cx channels in tumor outcome may be due, at least partially, to the fact that GJs not only interconnect cells from the same type, such as cancer cells, but also promote the intercellular communication of tumor cells with different types of cells from their microenvironment, and such diverse intercellular interactions have distinctive impact on tumor development. For example, whereas GJ-mediated interactions among tumor cells and microglia have been implicated in promotion of tumor growth, tumor cells delivery to dendritic cells of antigenic peptides through GJs have been associated with enhanced immune-mediated tumor elimination. In this review, we provide an updated overview on the role of GJICs in tumor immunity, focusing on the pro-tumor and antitumor effect of GJs occurring among tumor and immune cells. Accumulated data suggest that GJICs may act as tumor suppressors or enhancers depending on whether tumor cells interact predominantly with antitumor immune cells or with stromal cells. The complex modulation of immune-tumor cell GJICs should be taken into consideration in order to potentiate current cancer immunotherapies.
Collapse
Affiliation(s)
- María Alejandra Gleisner
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Mariela Navarrete
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Francisca Hofmann
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Andrés Tittarelli
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| |
Collapse
|
13
|
Mathews J, Levin M. Gap junctional signaling in pattern regulation: Physiological network connectivity instructs growth and form. Dev Neurobiol 2017; 77:643-673. [PMID: 27265625 PMCID: PMC10478170 DOI: 10.1002/dneu.22405] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 12/19/2022]
Abstract
Gap junctions (GJs) are aqueous channels that allow cells to communicate via physiological signals directly. The role of gap junctional connectivity in determining single-cell functions has long been recognized. However, GJs have another important role: the regulation of large-scale anatomical pattern. GJs are not only versatile computational elements that allow cells to control which small molecule signals they receive and emit, but also establish connectivity patterns within large groups of cells. By dynamically regulating the topology of bioelectric networks in vivo, GJs underlie the ability of many tissues to implement complex morphogenesis. Here, a review of recent data on patterning roles of GJs in growth of the zebrafish fin, the establishment of left-right patterning, the developmental dysregulation known as cancer, and the control of large-scale head-tail polarity, and head shape in planarian regeneration has been reported. A perspective in which GJs are not only molecular features functioning in single cells, but also enable global neural-like dynamics in non-neural somatic tissues has been proposed. This view suggests a rich program of future work which capitalizes on the rapid advances in the biophysics of GJs to exploit GJ-mediated global dynamics for applications in birth defects, regenerative medicine, and morphogenetic bioengineering. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 643-673, 2017.
Collapse
Affiliation(s)
- Juanita Mathews
- Department of Biology, Tufts Center for Regenerative and Developmental Biology, Tufts University, Medford, MA
| | - Michael Levin
- Department of Biology, Tufts Center for Regenerative and Developmental Biology, Tufts University, Medford, MA
| |
Collapse
|
14
|
Yu S, Xia M, Alsiddig MA, Liu H, Wei W, Chen J. Molecular cloning, alternative splicing and mRNA expression analysis of MAGI1 and its correlation with laying performance in geese. Br Poult Sci 2017; 58:158-165. [DOI: 10.1080/00071668.2016.1268251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- S. Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, PR China
- College of Life Science, Leshan Normal University, Sichuan, PR China
| | - M. Xia
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, PR China
| | - M. A. Alsiddig
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, PR China
| | - H. Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, PR China
| | - W. Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, PR China
| | - J. Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, PR China
| |
Collapse
|
15
|
Shen ZH, Zeng DF, Wang XY, Ma YY, Zhang X, Kong PY. Targeting of the leukemia microenvironment by c(RGDfV) overcomes the resistance to chemotherapy in acute myeloid leukemia in biomimetic polystyrene scaffolds. Oncol Lett 2016; 12:3278-3284. [PMID: 27899994 PMCID: PMC5103942 DOI: 10.3892/ol.2016.5042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 07/12/2016] [Indexed: 11/06/2022] Open
Abstract
The bone marrow microenvironment provides a relative sanctuary from cytotoxic drugs for leukemia cells. The present niche models concentrate on a two-dimensional (2D) co-culture system in vitro, which does not imitate the in vivo environment, while the 3D scaffolds are more reflective of this. Osteopontin (Opn) secreted by bone marrow osteoblasts, may participate in protecting leukemia cells from apoptosis by binding to its receptor αvβ3, which can be expressed on the surface of the leukemia MV4-11 cell line. However, the association between the Opn/αvβ3 axis and leukemia cells is unknown. In the present study, experiments were conducted on 3D polystyrene scaffolds coated with osteoblasts and leukemia cells. The cells were exposed to cyclo(Arg-Gly-Asp-d-Phe-Val) [c(RGDfV)] (35 nmol/ml), which blocks αvβ3, for a period of 24 h. Cytarabine was applied 24 h later. The adhesion, migration and apoptosis rates, and the cell cycle of the leukemia cells were analyzed after incubation for 24 and 48 h. In contrast to the 2D culture system, the stromal cells in the scaffolds secreted significantly more alkaline phosphatase and Opn (P<0.05). c(RGDfV) disrupted the adhesion and migration between the tumor cells and the matrix, induced the leukemia cells to leave the protective microenvironment and increased their sensitivity to cell cycle-dependent agents (P<0.05). In summary, the data certified that the 3D scaffolds are suitable for the growth of cells, and that c(RGDfV) inhibits the adhesion and migration abilities of leukemia cells in the endosteal niche. Therefore, blocking the function of Opn may be beneficial in the treatment of acute myeloid leukemia.
Collapse
Affiliation(s)
- Zhao-Hua Shen
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China; Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Dong-Feng Zeng
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Xiao-Yan Wang
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Ying-Ying Ma
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Xi Zhang
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Pei-Yan Kong
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
16
|
González-Nieto D, Chang KH, Fasciani I, Nayak R, Fernandez-García L, Barrio LC, Cancelas JA. Connexins: Intercellular Signal Transmitters in Lymphohematopoietic Tissues. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 318:27-62. [PMID: 26315883 DOI: 10.1016/bs.ircmb.2015.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Life-long hematopoietic demands are met by a pool of hematopoietic stem cells (HSC) with self-renewal and multipotential differentiation ability. Humoral and paracrine signals from the bone marrow (BM) hematopoietic microenvironment control HSC activity. Cell-to-cell communication through connexin (Cx) containing gap junctions (GJs) allows pluricellular coordination and synchronization through transfer of small molecules with messenger activity. Hematopoietic and surrounding nonhematopoietic cells communicate each other through GJs, which regulate fetal and postnatal HSC content and function in hematopoietic tissues. Traffic of HSC between peripheral blood and BM is also dependent on Cx proteins. Cx mutations are associated with human disease and hematopoietic dysfunction and Cx signaling may represent a target for therapeutic intervention. In this review, we illustrate and highlight the importance of Cxs in the regulation of hematopoietic homeostasis under normal and pathological conditions.
Collapse
Affiliation(s)
- Daniel González-Nieto
- Unit of Cellular and Animal Models, Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Kyung-Hee Chang
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Hoxworth Blood Center, University of Cincinnati, Cincinnati, OH, USA
| | - Ilaria Fasciani
- Unit of Experimental Neurology, Hospital Ramon y Cajal, Madrid, Spain
| | - Ramesh Nayak
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Laura Fernandez-García
- Unit of Cellular and Animal Models, Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
| | - Luis C Barrio
- Unit of Experimental Neurology, Hospital Ramon y Cajal, Madrid, Spain
| | - José A Cancelas
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Hoxworth Blood Center, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
17
|
Identification of Laying-Related SNP Markers in Geese Using RAD Sequencing. PLoS One 2015; 10:e0131572. [PMID: 26181055 PMCID: PMC4504669 DOI: 10.1371/journal.pone.0131572] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 06/03/2015] [Indexed: 12/18/2022] Open
Abstract
Laying performance is an important economical trait of goose production. As laying performance is of low heritability, it is of significance to develop a marker-assisted selection (MAS) strategy for this trait. Definition of sequence variation related to the target trait is a prerequisite of quantitating MAS, but little is presently known about the goose genome, which greatly hinders the identification of genetic markers for the laying traits of geese. Recently developed restriction site-associated DNA (RAD) sequencing is a possible approach for discerning large-scale single nucleotide polymorphism (SNP) and reducing the complexity of a genome without having reference genomic information available. In the present study, we developed a pooled RAD sequencing strategy for detecting geese laying-related SNP. Two DNA pools were constructed, each consisting of equal amounts of genomic DNA from 10 individuals with either high estimated breeding value (HEBV) or low estimated breeding value (LEBV). A total of 139,013 SNP were obtained from 42,291,356 sequences, of which 18,771,943 were for LEBV and 23,519,413 were for HEBV cohorts. Fifty-five SNP which had different allelic frequencies in the two DNA pools were further validated by individual-based AS-PCR genotyping in the LEBV and HEBV cohorts. Ten out of 55 SNP exhibited distinct allele distributions in these two cohorts. These 10 SNP were further genotyped in a goose population of 492 geese to verify the association with egg numbers. The result showed that 8 of 10 SNP were associated with egg numbers. Additionally, liner regression analysis revealed that SNP Record-111407, 106975 and 112359 were involved in a multiplegene network affecting laying performance. We used IPCR to extend the unknown regions flanking the candidate RAD tags. The obtained sequences were subjected to BLAST to retrieve the orthologous genes in either ducks or chickens. Five novel genes were cloned for geese which harbored the candidate laying-related SNP, including membrane associated guanylate kinase (MAGI-1), KIAA1462, Rho GTPase activating protein 21 (ARHGAP21), acyl-CoA synthetase family member 2 (ACSF2), astrotactin 2 (ASTN2). Collectively, our data suggests that 8 SNP and 5 genes might be promising candidate markers or targets for marker-assisted selection of egg numbers in geese.
Collapse
|
18
|
Harada T, Hirabayashi Y, Hatta Y, Tsuboi I, Glomm WR, Yasuda M, Aizawa S. Kinetics of hematopoietic stem cells and supportive activities of stromal cells in a three-dimensional bone marrow culture system. Growth Factors 2015; 33:347-55. [PMID: 26431462 DOI: 10.3109/08977194.2015.1088534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the bone marrow, hematopoietic cells proliferate and differentiate in close association with a three-dimensional (3D) hematopoietic microenvironment. Previously, we established a 3D bone marrow culture system. In this study, we analyzed the kinetics of hematopoietic cells, and more than 50% of hematopoietic progenitor cells, including CFU-Mix, CFU-GM and BFU-E in 3D culture were in a resting (non-S) phase. Furthermore, we examined the hematopoietic supportive ability of stromal cells by measuring the expression of various mRNAs relevant to hematopoietic regulation. Over the 4 weeks of culture, the stromal cells in the 3D culture are not needlessly activated and "quietly" regulate hematopoietic cell proliferation and differentiation during the culture, resulting in the presence of resting hematopoietic stem cells in the 3D culture for a long time. Thus, the 3D culture system may be a new tool for investigating hematopoietic stem cell-stromal cell interactions in vitro.
Collapse
Affiliation(s)
| | - Yukio Hirabayashi
- a Department of Functional Morphology and
- b Department of Medicine , Nihon University School of Medicine , Tokyo , Japan
| | - Yoshihiro Hatta
- b Department of Medicine , Nihon University School of Medicine , Tokyo , Japan
| | | | - Wilhelm Robert Glomm
- c Department of Chemical Engineering , Norwegian University of Science and Technology , Trondheim , Norway , and
| | - Masahiro Yasuda
- d Department of Chemical Engineering , Osaka Prefecture University , Osaka , Japan
| | | |
Collapse
|
19
|
Reikvam H, Ryningen A, Sæterdal LR, Nepstad I, Foss B, Bruserud Ø. Connexin expression in human acute myeloid leukemia cells: identification of patient subsets based on protein and global gene expression profiles. Int J Mol Med 2014; 35:645-52. [PMID: 25529637 PMCID: PMC4314410 DOI: 10.3892/ijmm.2014.2045] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/17/2014] [Indexed: 11/05/2022] Open
Abstract
Bone marrow stromal cells support both normal and malignant hematopoiesis. Τhis support is mediated through the local cytokine network and by direct cell‑cell interactions mediated via adhesion molecules and the formation of gap junctions by connexins. Previous studies on connexins in human acute myeloid leukemia (AML) have mainly focused on the investigation of leukemia cell lines. In the present study, we therefore investigated the expression of various connexins at the protein (i.e., cell surface expression) and mRNA level in primary human AML cells. The cell surface expression of the connexins, Cx26, Cx32, Cx37, Cx43 and Cx45, varied considerably between patients, and detectable levels were observed only for subsets of patients. On the whole, Cx43 and Cx45 showed the highest cell surface expression. Connexin expression was dependent on AML cell differentiation, but showed no association with cytogenetic abnormalities or mutations of the fms-related tyrosine kinase 3 (FLT3) or nucleophosmin (NPM)‑1 genes. By contrast, only Cx45 showed a significant variation between patients at the mRNA level. A high Cx45 expression was associated with the altered regulation of the mitogen‑activated protein kinase (MAPK) pathway and the release of pro-inflammatory cytokines [interleukin (IL)‑17, tumor necrosis factor (TNF), interferon‑γ], whereas a low Cx45 expression was associated with the altered regulation of protein functions (i.e., ligase activity, protein folding and catabolism). There was no significant correlation observed between the connexin mRNA and protein levels. Thus, differences in connexin expression can be used to subclassify AML patients. Differences in connexin cell surface expression profiles are not reflected at the mRNA level and have to be directly examined, whereas variations in Cx45 mRNA expression are associated with differences in cell signaling and the regulation of protein functions.
Collapse
Affiliation(s)
- Håkon Reikvam
- Institute of Clinical Science, University of Bergen, Bergen, Norway
| | - Anita Ryningen
- Institute of Clinical Science, University of Bergen, Bergen, Norway
| | - Lars Rune Sæterdal
- Department of Health Studies, University of Stavanger, Stravanger, Norway
| | - Ina Nepstad
- Institute of Clinical Science, University of Bergen, Bergen, Norway
| | - Brynjar Foss
- Department of Health Studies, University of Stavanger, Stravanger, Norway
| | - Øystein Bruserud
- Institute of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
20
|
Chicaybam L, Bonamino MH. Moving Receptor Redirected Adoptive Cell Therapy Toward Fine Tuning of Antitumor Responses. Int Rev Immunol 2014; 33:402-16. [DOI: 10.3109/08830185.2014.917412] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
21
|
Vignon C, Debeissat C, Georget MT, Bouscary D, Gyan E, Rosset P, Herault O. Flow cytometric quantification of all phases of the cell cycle and apoptosis in a two-color fluorescence plot. PLoS One 2013; 8:e68425. [PMID: 23935867 PMCID: PMC3728345 DOI: 10.1371/journal.pone.0068425] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 05/29/2013] [Indexed: 11/19/2022] Open
Abstract
An optimal technology for cell cycle analysis would allow the concomitant measurement of apoptosis, G0, G1, S, G2 and M phases in combination with cell surface phenotyping. We have developed an easy method in flow cytometry allowing this discrimination in an only two-color fluorescent plot. It is based on the concomitant use of 7-amino-actinomycin D and the antibodies anti-Ki67 and anti-phospho(Ser10)-histone H3, both conjugated to Alexa Fluor®488 to discriminate G0 and M phases, respectively. The method is particularly valuable in a clinical setting as verified in our laboratory by analyzing human leukemic cells from marrow samples or after exposure to cell cycle modifiers.
Collapse
Affiliation(s)
- Christine Vignon
- Université François-Rabelais de Tours, Tours, France
- CNRS, UMR 7292, LNOx team, Tours, France
| | | | | | - Didier Bouscary
- Institut Cochin, Université Paris Descartes, CNRS UMR 8104, Paris, France
| | - Emmanuel Gyan
- Université François-Rabelais de Tours, Tours, France
- CNRS, UMR 7292, LNOx team, Tours, France
- CHRU de Tours, Service d’Hématologie Clinique et Thérapie Cellulaire, Tours, France
| | - Philippe Rosset
- CHRU de Tours, Service de Chirurgie Orthopédique et Traumatologique, Tours, France
| | - Olivier Herault
- Université François-Rabelais de Tours, Tours, France
- CNRS, UMR 7292, LNOx team, Tours, France
- CHRU de Tours, Service d’Hématologie Biologique, Tours, France
- * E-mail:
| |
Collapse
|
22
|
Bischof AG, Yüksel D, Mammoto T, Mammoto A, Krause S, Ingber DE. Breast cancer normalization induced by embryonic mesenchyme is mediated by extracellular matrix biglycan. Integr Biol (Camb) 2013; 5:1045-56. [DOI: 10.1039/c3ib40103k] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Ashley G. Bischof
- Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA. Fax: +1 617-432-7828; Tel: +1 617-432-7044
- Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA
| | - Deniz Yüksel
- Wyss Institute for Biologically Inspired Engineering at Harvard University, 3 Blackfan Circle, CLSB 5th Floor, Boston, MA 02115, USA
| | - Tadanori Mammoto
- Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA. Fax: +1 617-432-7828; Tel: +1 617-432-7044
| | - Akiko Mammoto
- Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA. Fax: +1 617-432-7828; Tel: +1 617-432-7044
| | - Silva Krause
- Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA. Fax: +1 617-432-7828; Tel: +1 617-432-7044
| | - Donald E. Ingber
- Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA. Fax: +1 617-432-7828; Tel: +1 617-432-7044
- Wyss Institute for Biologically Inspired Engineering at Harvard University, 3 Blackfan Circle, CLSB 5th Floor, Boston, MA 02115, USA
- Harvard School of Engineering and Applied Sciences, Cambridge, MA 02138, USA
| |
Collapse
|
23
|
Paraguassú-Braga FH, Alves APG, Andrade Santos IMA, Bonamino M, Bonomo A. An Ectopic Stromal Implant Model for Hematopoietic Reconstitution and in Vivo Evaluation of Bone Marrow Niches. Cell Transplant 2012; 21:2677-88. [DOI: 10.3727/096368912x636993] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In adults, hematopoiesis takes places in the bone marrow, where specialized niches containing mesenchymal nonhematopoietic cells (stroma) harbor the hematopoietic stem cell (HSC). These niches are responsible and essential for the maintenance of HSCs. Attempts to expand HSCs fail to keep the general properties of stem cells, which depend on several niche components difficult to reproduce in in vitro culture systems. Here, we describe a methodology for in vivo study of hematopoietic stroma. We use stroma-loaded macroporous microcarriers implanted in the subcutaneous tissue of experimental animals and show that the ectopic stroma implant (ESI) is able to support hematopoiesis. Moreover, lethally irradiated mice can be rescued by ESI preloaded with HSCs, showing that they function as an ectopic bone marrow. ESI is also shown as a good system to study the role of different niche components. As an example, we used stromas lacking connexin 43 (Cx43) and confirm the importance of this molecule in the maintenance of the HSC niche in vivo. We believe ESI can work as an ectopic bone marrow allowing in vivo testing of different niches components and opening new avenues for the treatment of a variety of hematologic conditions particularly when stromal cell defects are the main cause of disease.
Collapse
Affiliation(s)
- Flávio Henrique Paraguassú-Braga
- Banco de Sangue de Cordão Umbilical e Placentário, Centro de Transplante de Medula Óssea, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Ana Paula G. Alves
- Programa de Medicina Experimental, Coordenação Geral Técnico-Científica, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | | | - Martin Bonamino
- Programa de Medicina Experimental, Coordenação Geral Técnico-Científica, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Adriana Bonomo
- Programa de Medicina Experimental, Coordenação Geral Técnico-Científica, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Hale JS, Li M, Lathia JD. The malignant social network: cell-cell adhesion and communication in cancer stem cells. Cell Adh Migr 2012; 6:346-55. [PMID: 22796941 DOI: 10.4161/cam.21294] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Tumors contain a vastly complicated cellular network that relies on local communication to execute malignant programs. The molecular cues that are involved in cell-cell adhesion orchestrate large-scale tumor behaviors such as proliferation and invasion. We have recently begun to appreciate that many tumors contain a high degree of cellular heterogeneity and are organized in a cellular hierarchy, with a cancer stem cell (CSC) population identified at the apex in multiple cancer types. CSCs reside in unique microenvironments or niches that are responsible for directing their behavior through cellular interactions between CSCs and stromal cells, generating a malignant social network. Identifying cell-cell adhesion mechanisms in this network has implications for the basic understanding of tumorigenesis and the development of more effective therapies. In this review, we will discuss our current understanding of cell-cell adhesion mechanisms used by CSCs and how these local interactions have global consequences for tumor biology.
Collapse
Affiliation(s)
- James S Hale
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | | | | |
Collapse
|
25
|
Sun J, Zheng Y, Feng X, Du W, Liu BF. Analysis of intercellular calcium signaling using microfluidic adjustable laminar flow for localized chemical stimulation. Anal Chim Acta 2012; 721:104-9. [PMID: 22405307 DOI: 10.1016/j.aca.2012.01.053] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Revised: 01/20/2012] [Accepted: 01/26/2012] [Indexed: 01/09/2023]
Abstract
The propagation of intercellular calcium signals provides a mechanism to coordinate cell population activity, which is essential for regulating cell behavior and organ development. However, existing analytical methods are difficult to realize localized chemical stimulation of a single cell among a population of cells that are in close contact with one another for studying the propagation of calcium wave. In this work, a microfluidic method is presented for the analysis of contact-dependent propagation of intercellular calcium wave induced by extracellular ATP using multiple laminar flows. Adjacent cells were seeded ∼300 μm downstream the intersection of a Y-shaped microchannel with negative pressure pulses. Consequently, the lateral diffusion distance of the chemical at cell locations was limited to ∼26 μm with a total flow rate of 20 μL min(-1), which prevented the interference of diffusion-induced cellular responses. Localized stimulation of the target cell with ATP induced the propagation of intercellular calcium wave among the cell population. In addition, studies on the spread of intercellular calcium wave under octanol inhibition allowed us to characterize the gap junction mediated cell-cell communication. Thus, this novel device will provide a versatile platform for intercellular signal transduction studies and high throughput drug screening.
Collapse
Affiliation(s)
- Jian Sun
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics, Department of Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, People's Republic of China
| | | | | | | | | |
Collapse
|
26
|
Acharya C, Adesida A, Zajac P, Mumme M, Riesle J, Martin I, Barbero A. Enhanced chondrocyte proliferation and mesenchymal stromal cells chondrogenesis in coculture pellets mediate improved cartilage formation. J Cell Physiol 2011; 227:88-97. [PMID: 22025108 DOI: 10.1002/jcp.22706] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In this study, we aimed at investigating the interactions between primary chondrocytes and mesenchymal stem/stromal cells (MSC) accounting for improved chondrogenesis in coculture systems. Expanded MSC from human bone marrow (BM-MSC) or adipose tissue (AT-MSC) were cultured in pellets alone (monoculture) or with primary human chondrocytes from articular (AC) or nasal (NC) cartilage (coculture). In order to determine the reached cell number and phenotype, selected pellets were generated by combining: (i) human BM-MSC with bovine AC, (ii) BM-MSC from HLA-A2+ with AC from HLA-A2- donors, or (iii) human green fluorescent protein transduced BM-MSC with AC. Human BM-MSC and AC were also cultured separately in transwells. Resulting tissues and/or isolated cells were assessed immunohistologically, biochemically, cytofluorimetrically, and by RT-PCR. Coculture of NC or AC (25%) with BM-MSC or AT-MSC (75%) in pellets resulted in up to 1.6-fold higher glycosaminoglycan content than what would be expected based on the relative percentages of the different cell types. This effect was not observed in the transwell model. BM-MSC decreased in number (about fivefold) over time and, if cocultured with chondrocytes, increased type II collagen and decreased type X collagen expression. Instead, AC increased in number (4.2-fold) if cocultured with BM-MSC and maintained a differentiated phenotype. Chondro-induction in MSC-chondrocyte coculture is a robust process mediated by two concomitant effects: MSC-induced chondrocyte proliferation and chondrocyte-enhanced MSC chondrogenesis. The identified interactions between progenitor and mature cell populations may lead to the efficient use of freshly harvested chondrocytes for ex vivo cartilage engineering or in situ cartilage repair.
Collapse
|
27
|
Hirabayashi Y, Hatta Y, Takeuchi J, Tsuboi I, Harada T, Ono K, Glomm WR, Yasuda M, Aizawa S. Novel three-dimensional long-term bone marrow culture system using polymer particles with grafted epoxy-polymer-chains supports the proliferation and differentiation of hematopoietic stem cells. Exp Biol Med (Maywood) 2011; 236:1342-50. [DOI: 10.1258/ebm.2011.011075] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hematopoiesis occurs in the bone marrow, where primitive hematopoietic cells proliferate and differentiate in close association with a three-dimensional (3D) hematopoietic microenvironment composed of stromal cells. We examined the hematopoietic supportive ability of stromal cells in a 3D culture system using polymer particles with grafted epoxy polymer chains. Umbilical cord blood-derived CD34+ cells were co-cultivated with MS-5 stromal cells. They formed a 3D structure in the culture dish in the presence of particles, and the total numbers of cells and the numbers of hematopoietic progenitor cells, including colony-forming unit (CFU)-Mix, CFU-granulocyte-macrophage, CFU-megakaryocyte and burst-forming unit-erythroid, were measured every seven days. The hematopoietic supportive activity of the 3D culture containing polymer particles and stromal cells was superior to that of 2D culture, and allowed the expansion and maintenance of hematopoietic progenitor cells for more than 12 weeks. Various types of hematopoietic cells, including granulocytes, macrophages and megakaryocytes at different maturation stages, appeared in the 3D culture, suggesting that the CD34+ cells were able to differentiate into a range of blood cell types. Morphological examination showed that MS-5 stromal cells grew on the surface of the particles and bridged the gaps between them to form a 3D structure. Hematopoietic cells slipped into the 3D layer and proliferated within it, relying on the presence of the MS-5 cells. These results suggest that this 3D culture system using polymer particles reproduced the hematopoietic phenomenon in vitro, and might thus provide a new tool for investigating hematopoietic stem cell–stromal cell interactions.
Collapse
Affiliation(s)
- Yukio Hirabayashi
- Department of Internal Medicine
- Department of Functional Morphology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo 173-8610
| | | | | | - Isao Tsuboi
- Department of Functional Morphology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo 173-8610
| | - Tomonori Harada
- Department of Functional Morphology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo 173-8610
| | - Kentaro Ono
- Department of Chemical Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Wilhelm Robert Glomm
- Department of Chemical Engineering, Norwegian University of Science and Technology, Sem Sælands vei 4, Trondheim N-7491, Norway
| | - Masahiro Yasuda
- Department of Chemical Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Shin Aizawa
- Department of Functional Morphology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo 173-8610
| |
Collapse
|
28
|
Decrock E, Vinken M, Bol M, D'Herde K, Rogiers V, Vandenabeele P, Krysko DV, Bultynck G, Leybaert L. Calcium and connexin-based intercellular communication, a deadly catch? Cell Calcium 2011; 50:310-21. [PMID: 21621840 DOI: 10.1016/j.ceca.2011.05.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 05/03/2011] [Accepted: 05/05/2011] [Indexed: 10/18/2022]
Abstract
Ca(2+) is known as a universal messenger mediating a wide variety of cellular processes, including cell death. In fact, this ion has been proposed as the 'cell death master', not only at the intracellular but also at the intercellular level. The most direct form of intercellular spread of cell death is mediated by gap junction channels. These channels have been shown to propagate cell death as well as cell survival signals between the cytoplasm of neighbouring cells, reflecting the dual role of Ca(2+) signals, i.e. cell death versus survival. Its precursor, the unopposed hemichannel (half of a gap junction channel), has recently joined in as a toxic pore connecting the intracellular with the extracellular environment and allowing the passage of a range of substances. The biochemical nature of the so-called intercellular cell death molecule, transferred through gap junctions or released/taken up via hemichannels, remains elusive but several studies pinpoint Ca(2+) itself or its messenger inositol trisphosphate as the responsible masters in crime. Although direct evidence is still lacking, indirect data including Ca(2+) involvement in intercellular communication and cell death, and effects of intercellular communication on intracellular Ca(2+) homeostasis, support this hypothesis. In addition, hemichannels and their molecular building blocks, connexin or pannexin proteins, may exert their effects on Ca(2+)-dependent cell death at the intracellular level, independently from their channel functions. This review provides a cutting edge overview of the current knowledge and underscores the intimate connection between intercellular communication, Ca(2+) signalling and cell death.
Collapse
Affiliation(s)
- Elke Decrock
- Department of Basic Medical Sciences - Physiology Group, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Asadi-Khiavi M, Hamzeiy H, Khani S, Nakhlband A, Barar J. Gap junctions: the claymore for cancerous cells. BIOIMPACTS : BI 2011; 1:113-9. [PMID: 23678415 DOI: 10.5681/bi.2011.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 07/17/2011] [Accepted: 07/20/2011] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Gap junctions play an important role in the cell proliferation in mammalian cells as well as carcinogenesis. However, there are controversial issues about their role in cancer pathogenesis. This study was designed to evaluate genotoxicity and cytotoxicity of Carbenoxolone (CBX) as a prototype of inter-cellular gap junction blocker in MCF7 and BT20 human breast cancer cells. METHODS The MCF7and BT20 human breast cancer cell lines were cultivated, and treated at designated confluency with different doses of CBX. Cellular cytotoxicity was examined using standard colorimetric assay associated with cell viability tests. Gene expression evaluation was carried out using real time polymerase chain reaction (PCR). RESULTS MCF7 and BT20 cells were significantly affected by CBX in a dose dependent manner in cell viability assays. Despite varying expression of genes, down regulation of pro- and anti-apoptotic genes was observed in these cells. CONCLUSION Based upon this investigation, it can be concluded that CBX could affect both low and high proliferative types of breast cancer cell lines and disproportionate down regulation of both pre- and anti-apoptotic genes may be related to interacting biomolecules, perhaps via gap junctions.
Collapse
Affiliation(s)
- Masoud Asadi-Khiavi
- Department of Pharmacology and Toxicology, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran ; Research Center for Pharmaceutical Nanotechnology, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran ; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | | |
Collapse
|
30
|
Kandouz M, Batist G. Gap junctions and connexins as therapeutic targets in cancer. Expert Opin Ther Targets 2010; 14:681-92. [PMID: 20446866 DOI: 10.1517/14728222.2010.487866] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
IMPORTANCE OF THE FIELD Connexins (Cxs) and gap junctional intercellular communications (GJICs) play roles in cancer development, growth and metastasis. Experimental studies suggest that targeting Cxs may be a novel technique, either to inhibit tumor cell growth directly or to sensitize to various therapies. AREAS COVERED IN THIS REVIEW A brief introduction to the role of Cxs in cancer. The focus is mainly on data available in the literature regarding therapeutic aspects. WHAT THE READER WILL GAIN This article reviews the various strategies that take advantage of gap junctions and connexins to eliminate cancer cells, including use of the bystander effect (BE) in gene therapy, the effect of connexins on chemosensitization, the role of apoptotic processes and interactions with the microenvironment. Attempts to restore connexin expression at the transcriptional and post-transcriptional levels are described, as well as promising strategies recently explored. The potential and limitations of the approaches are discussed. TAKE HOME MESSAGE Connexins have multiple facets, singly, in hemichannel complexes, in gap junctions or interacting with different proteins. The regulation of their expression is not fully resolved and selective manipulation of Cxs expression is therefore a challenge. Although the therapeutic potential of connexins is undeniable, more effort is needed to study the regulation and functions of these proteins.
Collapse
Affiliation(s)
- Mustapha Kandouz
- Wayne State University, Department of Pathology, 5101 Cass Avenue, Chemistry Building, Detroit, Michigan 48202, USA.
| | | |
Collapse
|
31
|
Weisberg E, Sattler M, Ray A, Griffin JD. Drug resistance in mutant FLT3-positive AML. Oncogene 2010; 29:5120-34. [DOI: 10.1038/onc.2010.273] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
32
|
Dillmann F, Veldwijk MR, Laufs S, Sperandio M, Calandra G, Wenz F, Zeller J, Fruehauf S. Plerixafor inhibits chemotaxis toward SDF-1 and CXCR4-mediated stroma contact in a dose-dependent manner resulting in increased susceptibility of BCR-ABL+ cell to Imatinib and Nilotinib. Leuk Lymphoma 2010; 50:1676-86. [PMID: 19657955 DOI: 10.1080/10428190903150847] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Despite Imatinib's remarkable success in chronic myelogenous leukemia treatment, monotherapy frequently causes resistance, underlining the rationale for combination chemotherapy. A potential approach would be interrupting the SDF-1/CXCR4 axis using the selective CXCR4 antagonist Plerixafor (previously AMD3100), as this axis has been reported to provide survival-enhancing effects to myeloid progenitor cells. By efficient CXCR4 blocking in the CXCR4(+)/BCR-ABL(+) cell line BV-173, plerixafor (1-100 muM) significantly inhibits SDF-1alpha-mediated chemotaxis and cell migration toward the murine stroma cell line FBMD-1. Furthermore, plerixafor also significantly (10-100 muM) increased the detachment rate of SDF-1-mediated/VCAM-1-associated cell adherence under shear stress. Using a stroma-dependent coculture assay, plerixafor sensitized BCR-ABL(+) cells toward tyrosine kinase inhibitor therapy. Because the level of cell killing nearly reached that of samples cultured without stroma, a cell-cell interaction disruption seems to improve the efficacy of BCR-ABL-targeting drugs. In addition, we could show that exposure of BCR-ABL(+) cells to Imatinib or Nilotinib induced an increase in surface CXCR4 expression. Our data suggest that for BCR-ABL(+) leukemia, the selective blocking of the SDF-1/CXCR4 axis by plerixafor is a potential mechanism to overcome the protective effect of the bone marrow environment, thereby increasing the therapeutic potency of anti-BCR-ABL drugs and the therapeutic window.
Collapse
Affiliation(s)
- Falk Dillmann
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Simultaneous activation of p53 and inhibition of XIAP enhance the activation of apoptosis signaling pathways in AML. Blood 2009; 115:306-14. [PMID: 19897582 DOI: 10.1182/blood-2009-03-212563] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Activation of p53 by murine double minute (MDM2) antagonist nutlin-3a or inhibition of X-linked inhibitor of apoptosis (XIAP) induces apoptosis in acute myeloid leukemia (AML) cells. We demonstrate that concomitant inhibition of MDM2 by nutlin-3a and of XIAP by small molecule antagonists synergistically induced apoptosis in p53 wild-type OCI-AML3 and Molm13 cells. Knockdown of p53 by shRNA blunted the synergy, and down-regulation of XIAP by antisense oligonucleotide (ASO) enhanced nutlin-3a-induced apoptosis, suggesting that the synergy was mediated by p53 activation and XIAP inhibition. This is supported by data showing that inhibition of both MDM2 and XIAP by their respective ASOs induced significantly more cell death than either ASO alone. Importantly, p53 activation and XIAP inhibition enhanced apoptosis in blasts from patients with primary AML, even when the cells were protected by stromal cells. Mechanistic studies demonstrated that XIAP inhibition potentiates p53-induced apoptosis by decreasing p53-induced p21 and that p53 activation enhances XIAP inhibition-induced cell death by promoting mitochondrial release of second mitochondria-derived activator of caspases (SMAC) and by inducing the expression of caspase-6. Because both XIAP and p53 are presently being targeted in ongoing clinical trials in leukemia, the combination strategy holds promise for expedited translation into the clinic.
Collapse
|
34
|
Wei Z, Chen N, Guo H, Wang X, Xu F, Ren Q, Lu S, Liu B, Zhang L, Zhao H. Bone marrow mesenchymal stem cells from leukemia patients inhibit growth and apoptosis in serum-deprived K562 cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2009; 28:141. [PMID: 19883517 PMCID: PMC2779804 DOI: 10.1186/1756-9966-28-141] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Accepted: 11/03/2009] [Indexed: 11/10/2022]
Abstract
BACKGROUND The regulation of growth and apoptosis in K562 cells by human bone marrow mesenchymal stem cells (MSCs) from leukemia patients was investigated. METHODS K562 cells were cocultured with leukemic MSCs under serum deprivation. Cell Counting Kit-8 (CCK-8), PI staining, Annexin V/PI binding and FACS assays were used to investigate cell proliferation, cell cycle status, and apoptosis of K562 cells cultures in the presence or absence of 10% serum. Western blotting was used to determine the levels of Akt, phosphorylated Akt (p-Akt), the BCL-2 family member Bad, and phosphorylated Bad (p-Bad) proteins in K562 cells after coculturing with MSCs. The effects of LY294002 (a specific inhibitor of PI3K) on protein expression were also determined. RESULTS K562 cell proliferation was inhibited by coculture with MSCs and the dominant cell cycle was the G0-G1 phase. The proportion of apoptotic K562 cells was decreased and the levels of p-Akt and p-Bad were upregulated after exposing K562 cells to MSCs. However, when LY294002 was used, p-Akt and p-Bad proteins inK562 cells showed a significant reduction, while no distinct variation was seen in the nonphosphorylated Akt and Bad protein levels. CONCLUSION Leukemic MSCs can inhibit K562 cell expansion and modulate the cell cycle to a state of relative quiescence. This allows the K562 cells to endure adverse conditions such as serum starvation. The PI3K-Akt-Bad signaling pathway may be involved in this antiapoptotic process via phosphorylation of the Akt and Bad proteins. Blocking MSC-induced transduction of the PI3K-Akt-Bad pathway may be a potential strategy for a targeted therapy to combat leukemia.
Collapse
Affiliation(s)
- Zhaohui Wei
- The Affiliated Hospital, North China Coal Medical college, Tanshan, Heibei, 063000, PR China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Foss B, Tronstad KJ, Bruserud Ø. Connexin-based signaling in acute myelogenous leukemia (AML). BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1798:1-8. [PMID: 19883623 DOI: 10.1016/j.bbamem.2009.10.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Revised: 09/15/2009] [Accepted: 10/26/2009] [Indexed: 10/20/2022]
Abstract
Normal and malignant hematopoiesis are regulated by intercellular communication in the hematopoietic microenvironments, and both soluble mediators as well as direct cell-cell contact play important functional roles. Gap junctions are complex membrane structures that transfer molecules between neighboring cells and thereby alter intracellular signaling and metabolism. The gap junction building blocks, the connexins, are also involved in gap junction-independent intercellular communication by forming hemichannels that transfer substances between the intra- and extracellular spaces. Connexins are furthermore involved in cell regulation as single molecules by modulating intracellular pathways and possibly gene transcription. The role of connexins in leukemogenesis and leukemic cell functions are not well characterized. In this review, we describe the known effects of gap junctions and connexins in acute myelogenous leukemia and the diverse potential of connexins in acute myelogenous leukemia chemosensitivity, intracellular signaling and cell death regulation.
Collapse
Affiliation(s)
- Brynjar Foss
- Department of Health Studies, University of Stavanger, Stavanger, Norway.
| | | | | |
Collapse
|
36
|
Abstract
Evidence is accumulating that some forms of cell death, like apoptosis, are not only governed by the complex interplay between extracellular and intracellular signals but are also strongly influenced by intercellular communicative networks. The latter is provided by arrays of channels consisting of connexin proteins, with gap junctions directly connecting the cytoplasm of neighboring cells and hemichannels positioned as pores that link the cytoplasm to the extracellular environment. The role of gap junctions in cell death communication has received considerable interest and recently hemichannels have joined in as potentially toxic pores adding their part to the cell death process. However, despite a large body of existing evidence, especially for gap junctions, the exact contribution of the connexin channel family still remains controversial, as both gap junctions and hemichannels may furnish cell death as well as cell survival signals. An additional layer of complexity is formed by the fact that connexin proteins as such, beyond their channel function, may influence the cell death process. We here review the current knowledge on connexins and their channels in cell death and specifically address the molecular mechanisms that underlie connexin-related signaling. We also briefly focus on pannexins, a novel set of connexin-like proteins that have been implicated in cellular responses to pathological insults.
Collapse
|
37
|
Paredes-Gamero EJ, Leon CMMP, Borojevic R, Oshiro MEM, Ferreira AT. Changes in intracellular Ca2+ levels induced by cytokines and P2 agonists differentially modulate proliferation or commitment with macrophage differentiation in murine hematopoietic cells. J Biol Chem 2008; 283:31909-19. [PMID: 18775989 DOI: 10.1074/jbc.m801990200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The role of intracellular Ca2+ (Ca2+i) on hematopoiesis was investigated in long term bone marrow cultures using cytokines and agonists of P2 receptors. Cytokines interleukin 3 and granulocyte/macrophage colony stimulator factor promoted a modest increase in Ca2+i concentration ([Ca2+]i) with activation of phospholipase Cgamma, MEK1/2, and Ca2+/calmodulin kinase II. Involvement of protein kinase C was restricted to stimulation with interleukin 3. In addition, these cytokines promoted proliferation (20 times) and an increase in the Gr-1(-)Mac-1+ population with participation of gap junctions (GJ). Nevertheless ATP, ADP, and UTP promoted a large increase in [Ca2+]i, moderate proliferation (6 times), a reduction in the primitive Gr-1(-)Mac-1(-)c-Kit+ population, and differentiation into macrophages without participation of GJ. It is likely that Ca2+i participates as a regulator of hematopoietic signaling: moderate increases in [Ca2+]i would be related to cytokine-dependent proliferation with participation of GJ, whereas high increases in [Ca2+]i would be related to macrophage differentiation without maintenance of the primitive population.
Collapse
Affiliation(s)
- Edgar J Paredes-Gamero
- Department of Biophysics, Federal University of São Paulo, Rua Botucatu 862, 04023-062 São Paulo, Brazil.
| | | | | | | | | |
Collapse
|
38
|
Advantages and limitations of commonly used methods to assay the molecular permeability of gap junctional intercellular communication. Biotechniques 2008; 45:33-52, 56-62. [PMID: 18611167 DOI: 10.2144/000112810] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The role of gap junctional intercellular communication (GJIC) in regulation of normal growth and differentiation is becoming increasingly recognized as a major cellular function. GJIC consists of intercellular exchange of low molecular weight molecules, and is the only means for direct contact between cytoplasms of adjacent animal cells. Disturbances of GJIC have been associated with many pathological conditions, such as carcinogenesis or hereditary illness. Reliable and accurate methods for the determination of GJIC are therefore important in cell biology studies. There are several methods used successfully in numerous laboratories to measure GJIC both in vitro and in vivo. This review comments on techniques currently used to study cell-to-cell communication, either by measuring dye transfer, as in methods like microinjection, scrape loading, gap-fluorescence recovery after photobleaching (gap-FRAP), the preloading assay, and local activation of a molecular fluorescent probe (LAMP), or by measuring electrical conductance and metabolic cooperation. As we will discuss in this review, these techniques are not equivalent but instead provide complementary information. We will focus on their main advantages and limitations. Although biological applications guide the choice of techniques we describe, we also review points that must be taken into consideration before using a methodology, such as the number of cells to analyze.
Collapse
|
39
|
Weisberg E, Wright RD, McMillin DW, Mitsiades C, Ray A, Barrett R, Adamia S, Stone R, Galinsky I, Kung AL, Griffin JD. Stromal-mediated protection of tyrosine kinase inhibitor-treated BCR-ABL-expressing leukemia cells. Mol Cancer Ther 2008; 7:1121-9. [PMID: 18445657 DOI: 10.1158/1535-7163.mct-07-2331] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Clinical studies of patients with chronic myeloid leukemia revealed that a common pattern of response is a dramatic fall in the circulating population of blast cells, with a minimal or delayed decrease in marrow blasts, suggesting a protective environment. These observations suggest that a greater understanding of the interaction of stromal cells with leukemic cells is essential. Here, we present an in vivo system for monitoring relative tumor accumulation in leukemic mice and residual disease in leukemic mice treated with a tyrosine kinase inhibitor and an in vitro system for identifying integral factors involved in stromal-mediated cytoprotection. Using the in vivo model, we observed high tumor burden/residual disease in tissues characterized as significant sources of hematopoiesis-promoting stroma, with bone marrow stroma most frequently showing the highest accumulation of leukemia in untreated and nilotinib-treated mice as well as partial protection of leukemic cells from the inhibitory effects of nilotinib. These studies, which showed a pattern of leukemia distribution consistent with what is observed in imatinib- and nilotinib-treated chronic myeloid leukemia patients, were followed by a more in-depth analysis of stroma-leukemia cell interactions that lead to protection of leukemia cells from nilotinib-induced cytotoxicity. For the latter, we used the human BCR-ABL-positive cell line, KU812F, and the human bone marrow stroma cell line, HS-5, to more closely approximate the bone marrow-associated cytoprotection observed in drug-treated leukemia patients. This in vitro system helped to elucidate stromal-secreted viability factors that may play a role in stromal-mediated cytoprotection of tyrosine kinase inhibitor-treated leukemia cells.
Collapse
Affiliation(s)
- Ellen Weisberg
- Department of Medical Oncology/Hematologic Neoplasia, Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Effects of human bone marrow stromal cell line (HFCL) on the proliferation, differentiation and apoptosis of acute myeloid leukemia cell lines U937, HL-60 and HL-60/VCR. Int J Hematol 2008; 87:152-166. [DOI: 10.1007/s12185-008-0030-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2007] [Accepted: 11/05/2007] [Indexed: 10/22/2022]
|
42
|
Ong V, Liem NLM, Schmid MA, Verrills NM, Papa RA, Marshall GM, Mackenzie KL, Kavallaris M, Lock RB. A role for altered microtubule polymer levels in vincristine resistance of childhood acute lymphoblastic leukemia xenografts. J Pharmacol Exp Ther 2007; 324:434-42. [PMID: 17986648 DOI: 10.1124/jpet.107.128926] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The microtubule-depolymerizing drug, vincristine, is effective in the treatment of acute lymphoblastic leukemia (ALL). Although vincristine resistance mechanisms have been extensively characterized in cell lines, their clinical relevance is poorly understood. The aim of the current study was to define clinically relevant mechanisms of vincristine resistance in a panel of childhood ALL xenografts established in immune-deficient (nonobese diabetic/severe combined immunodeficient) mice. We also studied two independent xenograft sublines that were selected by in vivo vincristine exposure. In vitro vincristine sensitivity determined by a stromal coculture, murine bone marrow stromal cell line (MS-5), assay, but not methyl-thiazolyl-tetrazolium metabolic activity assay, significantly correlated (P = 0.05) with the length of the patients' first remission. Investigations into mechanisms of resistance revealed no association with steady-state vincristine accumulation or increased activity and/or expression of ATP-binding cassette transporters, although increased intracellular levels of polymerized tubulin significantly correlated with resistance (r = 0.85; P = 0.0019). Two xenograft sublines selected by in vivo vincristine exposure exhibited a 2-fold increase in polymerized tubulin levels compared with the parental subline (P < 0.05), reflecting their in vivo vincristine resistance. In this study, a vincristine-resistant xenograft with high levels of polymerized tubulin was relatively sensitive to the microtubule-polymerizing drug paclitaxel. These results indicate that the balance between polymerized and nonpolymerized tubulin may be an important determinant of response to Vinca alkaloid-based chemotherapy regimens in childhood ALL.
Collapse
Affiliation(s)
- Vivienna Ong
- Children's Cancer Institute Australia, PO Box 81, High St., Randwick 2031, NSW, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Levin M. Gap junctional communication in morphogenesis. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2007; 94:186-206. [PMID: 17481700 PMCID: PMC2292839 DOI: 10.1016/j.pbiomolbio.2007.03.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Gap junctions permit the direct passage of small molecules from the cytosol of one cell to that of its neighbor, and thus form a system of cell-cell communication that exists alongside familiar secretion/receptor signaling. Because of the rich potential for regulation of junctional conductance, and directional and molecular gating (specificity), gap junctional communication (GJC) plays a crucial role in many aspects of normal tissue physiology. However, the most exciting role for GJC is in the regulation of information flow that takes place during embryonic development, regeneration, and tumor progression. The molecular mechanisms by which GJC establishes local and long-range instructive morphogenetic cues are just beginning to be understood. This review summarizes the current knowledge of the involvement of GJC in the patterning of both vertebrate and invertebrate systems and discusses in detail several morphogenetic systems in which the properties of this signaling have been molecularly characterized. One model consistent with existing data in the fields of vertebrate left-right patterning and anterior-posterior polarity in flatworm regeneration postulates electrophoretically guided movement of small molecule morphogens through long-range GJC paths. The discovery of mechanisms controlling embryonic and regenerative GJC-mediated signaling, and identification of the downstream targets of GJC-permeable molecules, represent exciting next areas of research in this fascinating field.
Collapse
Affiliation(s)
- Michael Levin
- Forsyth Center for Regenerative and Devlopmental Biology, Forsyth Institute, and Developmental Biology Department, Harvard School of Dental Medicine, Boston, MA 02115, USA.
| |
Collapse
|
44
|
Esser AT, Smith KC, Weaver JC, Levin M. Mathematical model of morphogen electrophoresis through gap junctions. Dev Dyn 2006; 235:2144-59. [PMID: 16786594 DOI: 10.1002/dvdy.20870] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Gap junctional communication is important for embryonic morphogenesis. However, the factors regulating the spatial properties of small molecule signal flows through gap junctions remain poorly understood. Recent data on gap junctions, ion transporters, and serotonin during left-right patterning suggest a specific model: the net unidirectional transfer of small molecules through long-range gap junctional paths driven by an electrophoretic mechanism. However, this concept has only been discussed qualitatively, and it is not known whether such a mechanism can actually establish a gradient within physiological constraints. We review the existing functional data and develop a mathematical model of the flow of serotonin through the early Xenopus embryo under an electrophoretic force generated by ion pumps. Through computer simulation of this process using realistic parameters, we explored quantitatively the dynamics of morphogen movement through gap junctions, confirming the plausibility of the proposed electrophoretic mechanism, which generates a considerable gradient in the available time frame. The model made several testable predictions and revealed properties of robustness, cellular gradients of serotonin, and the dependence of the gradient on several developmental constants. This work quantitatively supports the plausibility of electrophoretic control of morphogen movement through gap junctions during early left-right patterning. This conceptual framework for modeling gap junctional signaling -- an epigenetic patterning mechanism of wide relevance in biological regulation -- suggests numerous experimental approaches in other patterning systems.
Collapse
Affiliation(s)
- Axel T Esser
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | | | | |
Collapse
|
45
|
Fonseca PC, Nihei OK, Savino W, Spray DC, Alves LA. Flow cytometry analysis of gap junction-mediated cell-cell communication: advantages and pitfalls. Cytometry A 2006; 69:487-93. [PMID: 16646046 DOI: 10.1002/cyto.a.20255] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Since the first morphological description of the gap junctions use electron microscopy, a considerable number of techniques has been introduced to evaluate gap junction channel functionality, many of which use dye transfer techniques, such as dye injection and fluorescent dye transfer, analyzed by flow cytometry. METHODS To analyze dye transfer, generally one population of cells is incubated with calcein-AM (0.5 microM) for 30 min at 37 degrees C, and the other population was incubated with the lipophilic dye DiIC(18) (3) (10 microM) for 1 h at 37 degrees C; after incubation, these cells were washed five times with PBS and cocultured for different times, and then the dye transfer was analyzed by flow cytometry. RESULTS In this short overview, we focus on some advantages and disadvantages of flow cytometry as a technique to investigate gap junction-mediated intercellular communication (GJIC). In addition, we point out some technical pitfalls that we have encountered when applying this technique to study gap junctions in immune system cells. CONCLUSIONS Analysis of fluorescent dye transfer by flow cytometry is a useful tool to investigate GJIC. However, some points must be taken into consideration before using this methodology, which are discussed herein.
Collapse
Affiliation(s)
- Paula Candida Fonseca
- Laboratório de Comunicação Celular, Departamento de Imunologia, Instituto Oswaldo Cruz, Fundação, Rio de Janeiro, Brasil
| | | | | | | | | |
Collapse
|
46
|
Study on the bone marrow mesenchymal stem cells induced drug resistance in the U937 cells and its mechanism. Chin Med J (Engl) 2006. [DOI: 10.1097/00029330-200606010-00005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
47
|
Abstract
This review discusses current understanding of the role that endogenous and exogenous progenitor cells may have in the treatment of the diseased heart. In the last several years, a major effort has been made in an attempt to identify immature cells capable of differentiating into cell lineages different from the organ of origin to be employed for the regeneration of the damaged heart. Embryonic stem cells (ESCs) and bone marrow-derived cells (BMCs) have been extensively studied and characterized, and dramatic advances have been made in the clinical application of BMCs in heart failure of ischemic and nonischemic origin. However, a controversy exists concerning the ability of BMCs to acquire cardiac cell lineages and reconstitute the myocardium lost after infarction. The recognition that the adult heart possesses a stem cell compartment that can regenerate myocytes and coronary vessels has raised the unique possibility to rebuild dead myocardium after infarction, to repopulate the hypertrophic decompensated heart with new better functioning myocytes and vascular structures, and, perhaps, to reverse ventricular dilation and wall thinning. Cardiac stem cells may become the most important cell for cardiac repair.
Collapse
Affiliation(s)
- Annarosa Leri
- Cardiovascular Research Institute, Department of Medicine, New York Medical College, Valhalla, NY10595, USA
| | | | | |
Collapse
|
48
|
Leibundgut K, Schmitz NMR, Hirt A. Catalytic Activities of G1Cyclin-Dependent Kinases and Phosphorylation of Retinoblastoma Protein in Mobilized Peripheral Blood CD34+Hematopoietic Progenitor Cells. Stem Cells 2005; 23:1002-11. [PMID: 15941859 DOI: 10.1634/stemcells.2004-0088] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Depending on the source of cells, the cell cycle status of hematopoietic stem and progenitor cells capable of repopulating the marrow of transplant recipients is controversial. In this study, using biochemical methods, the cell cycle status of mobilized CD34+ cells was analyzed. It was demonstrated in CD34+ cell extracts that there was high catalytic activity of G(1) cyclin-dependent kinases 4 and 6 (CDK4 and CDK6) but low activity of CDK2. This was in contrast to the resting reference cells that showed only minimal or no activity of these CDKs. Since at the G0-->G1-->S transition CDK4/6 and CDK2 sequentially phosphorylate the retinoblastoma protein (pRB), its phosphorylation status was analyzed. Previously, we showed that p110RB was unphosphorylated at serine (Ser)-608 in CD34+ cells, consistent with the ability to suppress cell growth. Here, it was established that this form of pRB was phosphorylated at Ser-780, Ser-795, and Ser-807/811 in CD34+ but not in resting reference cells. This result was therefore consistent with the presence of high CDK4/6 activities in CD34+ cells. Conversely, CDK2 activity was low and the pRB residues Ser-612 and threonine (Thr)-821, which are exclusively phosphorylated by CDK2 in conjunction with either cyclin E or A, were unphosphorylated in >90% of CD34+ cells. We therefore show for the first time the exact position of mobilized CD34+ cells within the cell cycle; that is, they do not reside in G0 but in early G1 phase and did not cross the restriction point into late G1 phase.
Collapse
Affiliation(s)
- Kurt Leibundgut
- Department of Pediatrics, University of Bern Inselspital, CH-3010, Bern, Switzerland.
| | | | | |
Collapse
|
49
|
Trosko JE, Upham BL. The emperor wears no clothes in the field of carcinogen risk assessment: ignored concepts in cancer risk assessment. Mutagenesis 2005; 20:81-92. [PMID: 15784692 DOI: 10.1093/mutage/gei017] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The following is a position paper challenging the paradigm that 'carcinogen = mutagen', and that the current rodent bioassay to predict risks to human cancers is relevant and useful. Specifically, we review current observations concerning carcinogenesis that might lead to another approach for assessing the identification of human carcinogenic hazards and the risk assessment that chemicals might pose. We give a brief review of the multistage and multimechanism process of cancer in a tissue that involves not only genotoxic but also epigenetic events, and the importance of stem and progenitor cells in the development of cancer. We focus on the often ignored 'epigenetic' effects of carcinogens and the role of cell communication systems in epigenetically altering gene expression that leads to an imbalance of cell proliferation, differentiation and apoptosis in a tissue that can contribute to the cancer process. To draw attention to the fact that the current paradigm and policy to test toxic chemicals is often misleading and incorrect, we discuss how oxidative stress, in spite of the DNA damaging data, most probably contributes to cancer at the epigenetic level. Additionally, we briefly review how this mutagenic concept has greatly diverted attention away from doing research on the lower molecular weight, non-genotoxic, polycyclic aromatic hydrocarbons (PAHs), and how these low molecular weight PAHs are etiologically more relevant to the disease potential of environmental mixtures such as cigarette smoke.
Collapse
Affiliation(s)
- James E Trosko
- National Food Safety Toxicology Center, Department of Pediatrics and Human Development, Michigan State University, East Lansing, MI 48824, USA.
| | | |
Collapse
|