1
|
Torres-Machorro AL, Becerril C, Hernández-Plata E, Luis-García ER, Maldonado M, Herrera I, Negreros M, Hernández-Sánchez F, Mendoza-Milla C, Gaxiola M, Ramírez R, Pardo A, Buendía-Roldán I, Selman M, Cisneros J. Altered expression pattern of immune response-related genes and isoforms in hypersensitivity pneumonitis lung fibroblasts. Sci Rep 2024; 14:24002. [PMID: 39402115 PMCID: PMC11473681 DOI: 10.1038/s41598-024-74267-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/24/2024] [Indexed: 10/17/2024] Open
Abstract
Hypersensitivity pneumonitis (HP) is an immune-mediated inflammatory interstitial lung disease that may evolve to pulmonary fibrosis, a progressive disorder with a poor prognosis characterized by fibroblast activation and extracellular matrix accumulation. In HP lung fibroblasts, the gene expression of proteins involved in the interaction with the immune response, their isoforms, and how they influence their phenotype have yet to be elucidated. We analyzed the expression and splicing variants of 16 target genes involved in the interaction between HP fibroblasts and immune signaling and evaluated possible correlations with clinical data. The comparison of HP and control fibroblasts revealed distinct gene expression patterns. HP lung fibroblasts displayed an increased expression of IFI27 and PDFGRA and a downregulation of IL17RC and TGFBR3. IFI27 immunoreactive protein was markedly increased in HP lung tissues and normal fibroblasts treated with TGF-β. Furthermore, IFI27 overexpression in normal fibroblasts increased α-SMA and decreased cell number over time. The isoform analysis showed similar expression patterns for most genes, except for the AGER receptor with increased soluble variants relative to full-length AGER in HP fibroblasts. These findings indicate important differences in the expression of genes related to the immune response by HP fibroblasts, highlighting their unique characteristics and providing further insight into a possible profibrotic role of IFI27 in the disease.
Collapse
Affiliation(s)
- Ana Lilia Torres-Machorro
- Laboratorio de Biología Celular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Carina Becerril
- Laboratorio de Biología Celular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Everardo Hernández-Plata
- Investigador Por México, Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCyT), and Instituto Nacional de Medicina Genómica, 14610, Ciudad de México, México
| | - Erika Rubí Luis-García
- Laboratorio de Biología Celular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Mariel Maldonado
- Laboratorio de Biopatología Pulmonar INER-Ciencias-UNAM, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Iliana Herrera
- Laboratorio de Biopatología Pulmonar INER-Ciencias-UNAM, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Miguel Negreros
- Clínica de Vasculitis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Fernando Hernández-Sánchez
- Departamento de Investigación en Virología y Micología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Criselda Mendoza-Milla
- Laboratorio de Transducción de Señales, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Miguel Gaxiola
- Laboratorio de Morfología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Remedios Ramírez
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Annie Pardo
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Ivette Buendía-Roldán
- Laboratorio de Investigación Traslacional en Envejecimiento y Enfermedades Fibrosantes, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Moisés Selman
- Laboratorio de Biopatología Pulmonar INER-Ciencias-UNAM, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - José Cisneros
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México.
| |
Collapse
|
2
|
Marchand T, Pinho S. Leukemic Stem Cells: From Leukemic Niche Biology to Treatment Opportunities. Front Immunol 2021; 12:775128. [PMID: 34721441 PMCID: PMC8554324 DOI: 10.3389/fimmu.2021.775128] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/28/2021] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is one of the most common types of leukemia in adults. While complete remission can be obtained with intensive chemotherapy in young and fit patients, relapse is frequent and prognosis remains poor. Leukemic cells are thought to arise from a pool of leukemic stem cells (LSCs) which sit at the top of the hierarchy. Since their discovery, more than 30 years ago, LSCs have been a topic of intense research and their identification paved the way for cancer stem cell research. LSCs are defined by their ability to self-renew, to engraft into recipient mice and to give rise to leukemia. Compared to healthy hematopoietic stem cells (HSCs), LSCs display specific mutations, epigenetic modifications, and a specific metabolic profile. LSCs are usually considered resistant to chemotherapy and are therefore the drivers of relapse. Similar to their HSC counterpart, LSCs reside in a highly specialized microenvironment referred to as the “niche”. Bidirectional interactions between leukemic cells and the microenvironment favor leukemic progression at the expense of healthy hematopoiesis. Within the niche, LSCs are thought to be protected from genotoxic insults. Improvement in our understanding of LSC gene expression profile and phenotype has led to the development of prognosis signatures and the identification of potential therapeutic targets. In this review, we will discuss LSC biology in the context of their specific microenvironment and how a better understanding of LSC niche biology could pave the way for new therapies that target AML.
Collapse
Affiliation(s)
- Tony Marchand
- Service d'Hématologie Clinique, Centre Hospitalier Universitaire de Rennes, Rennes, France.,Faculté de médecine, Université Rennes 1, Rennes, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U1236, Rennes, France
| | - Sandra Pinho
- Department of Pharmacology & Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
3
|
Bolandi SM, Pakjoo M, Beigi P, Kiani M, Allahgholipour A, Goudarzi N, Khorashad JS, Eiring AM. A Role for the Bone Marrow Microenvironment in Drug Resistance of Acute Myeloid Leukemia. Cells 2021; 10:2833. [PMID: 34831055 PMCID: PMC8616250 DOI: 10.3390/cells10112833] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with a poor prognosis and remarkable resistance to chemotherapeutic agents. Understanding resistance mechanisms against currently available drugs helps to recognize the therapeutic obstacles. Various mechanisms of resistance to chemotherapy or targeted inhibitors have been described for AML cells, including a role for the bone marrow niche in both the initiation and persistence of the disease, and in drug resistance of the leukemic stem cell (LSC) population. The BM niche supports LSC survival through direct and indirect interactions among the stromal cells, hematopoietic stem/progenitor cells, and leukemic cells. Additionally, the BM niche mediates changes in metabolic and signal pathway activation due to the acquisition of new mutations or selection and expansion of a minor clone. This review briefly discusses the role of the BM microenvironment and metabolic pathways in resistance to therapy, as discovered through AML clinical studies or cell line and animal models.
Collapse
Affiliation(s)
- Seyed Mohammadreza Bolandi
- Department of Immunology, Razi Vaccine and Sera Research Institute, Karaj, Iran; (S.M.B.); (N.G.)
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Mahdi Pakjoo
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; (M.P.); (P.B.)
| | - Peyman Beigi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; (M.P.); (P.B.)
| | - Mohammad Kiani
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Ali Allahgholipour
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Negar Goudarzi
- Department of Immunology, Razi Vaccine and Sera Research Institute, Karaj, Iran; (S.M.B.); (N.G.)
| | - Jamshid S. Khorashad
- Centre for Haematology, Hammersmith Hospital, Imperial College London, London W12 0HS, UK;
| | - Anna M. Eiring
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
| |
Collapse
|
4
|
Filatova EN, Sakharnov NA, Knyazev DI, Utkin OV. Changes in mRNA expression of members of TGFB1-associated pathways in human leukocytes during EBV infection. Acta Microbiol Immunol Hung 2019; 66:247-254. [PMID: 30465450 DOI: 10.1556/030.65.2018.047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Transforming growth factor β 1 (TGFB1) likely contributes to the pathogenesis of Epstein-Barr virus (EBV)-mediated cancer. A microarray containing 59 probes for detecting mRNA of members of TGFB1-associated pathways was developed. mRNA expression of TGFB1 receptors and members of connected pathways were examined in peripheral blood leukocytes of patients during acute EBV infection and after recovery. TGFB1 and TGFBR2 mRNA expression was increased in patients with EBV infection. Similarly, mRNA expression of protein kinase C (PRKCB), MAP3K7, PDLIM7, and other members of TGFB1 and NF-κB signaling pathways increased. A shift of mRNA transcript variant expression of some key members (TGFBR2, PRKCB, and NFKBIB) of involved signaling pathways was detected. After the patients' recovery, most of the altered mRNA expression has been normalized. We speculate that in patients with EBV infection, members of TGFB1-associated pathways contribute to the suppression of proapoptotic and induction of pro-survival factors in leukocytes. The modulation of TGFB1-associated pathways may be considered as a potential risk factor in the development of EBV-associated tumors in patients with acute EBV infection.
Collapse
Affiliation(s)
- Elena Nikolaevna Filatova
- 1 Laboratory of Molecular Biology and Biotechnology, Blokhina Scientific Research Institute of Epidemiology and Microbiology of Nizhny Novgorod , Nizhny Novgorod, Russian Federation
| | - Nikolay Aleksandrovich Sakharnov
- 1 Laboratory of Molecular Biology and Biotechnology, Blokhina Scientific Research Institute of Epidemiology and Microbiology of Nizhny Novgorod , Nizhny Novgorod, Russian Federation
| | - Dmitry Igorevich Knyazev
- 1 Laboratory of Molecular Biology and Biotechnology, Blokhina Scientific Research Institute of Epidemiology and Microbiology of Nizhny Novgorod , Nizhny Novgorod, Russian Federation
| | - Oleg Vladimirovich Utkin
- 1 Laboratory of Molecular Biology and Biotechnology, Blokhina Scientific Research Institute of Epidemiology and Microbiology of Nizhny Novgorod , Nizhny Novgorod, Russian Federation
| |
Collapse
|
5
|
Bjelica S, Diklić M, Đikić D, Kovačić M, Subotički T, Mitrović-Ajtić O, Radojković M, Čokić VP, Santibanez JF. Hydroxyurea-induced senescent peripheral blood mesenchymal stromal cells inhibit bystander cell proliferation of JAK2V617F-positive human erythroleukemia cells. FEBS J 2019; 286:3647-3663. [PMID: 31090259 DOI: 10.1111/febs.14927] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 04/15/2019] [Accepted: 05/13/2019] [Indexed: 12/16/2022]
Abstract
Hydroxyurea (HU) is a nonalkylating antineoplastic agent used in the treatment of hematological malignancies. HU is a DNA replication stress inducer, and as such, it may induce a premature senescence-like cell phenotype; however, its repercussion on bystander cell proliferation has not been revealed so far. Our results indicate that HU strongly inhibited peripheral blood mesenchymal stromal cells (PBMSC) proliferation by cell cycle arrest in S phase, and that, consequently, PBMSC acquire senescence-related phenotypical changes. HU-treated PBMSC display increased senescence-associated β-galactosidase levels and p16INK4 expression, as well as DNA damage response and genotoxic effects, evidenced by expression of γH2A.X and micronuclei. Moreover, HU-induced PBMSC senescence is mediated by increased reactive oxygen species (ROS) levels, as demonstrated by the inhibition of senescence markers in the presence of ROS scavenger N-acetylcysteine and NADPH oxidase inhibitor Apocynin. To determine the HU-induced bystander effect, we used the JAK2V617F-positive human erythroleukemia 92.1.7 (HEL) cells. Co-culture with HU-induced senescent PBMSC (HU-S-PBMSC) strongly inhibited bystander HEL cell proliferation, and this effect is mediated by both ROS and transforming growth factor (TGF)-β expression. Besides induction of premature senescence, HU educates PBMSC toward an inhibitory phenotype of HEL cell proliferation. Finally, our study contributes to the understanding of the role of HU-induced PBMSC senescence as a potential adjuvant in hematological malignancy therapies.
Collapse
Affiliation(s)
- Sunčica Bjelica
- Group for Molecular Oncology, Institute for Medical Research, University of Belgrade, Serbia
| | - Miloš Diklić
- Group for Molecular Oncology, Institute for Medical Research, University of Belgrade, Serbia
| | - Dragoslava Đikić
- Group for Molecular Oncology, Institute for Medical Research, University of Belgrade, Serbia
| | - Marijana Kovačić
- Group for Molecular Oncology, Institute for Medical Research, University of Belgrade, Serbia
| | - Tijana Subotički
- Group for Molecular Oncology, Institute for Medical Research, University of Belgrade, Serbia
| | - Olivera Mitrović-Ajtić
- Group for Molecular Oncology, Institute for Medical Research, University of Belgrade, Serbia
| | - Milica Radojković
- Department of Haematology, Clinical Hospital Centre Dragisa Misovic, Belgrade, Serbia.,Faculty of Medicine, University of Belgrade, Serbia
| | - Vladan P Čokić
- Group for Molecular Oncology, Institute for Medical Research, University of Belgrade, Serbia
| | - Juan F Santibanez
- Group for Molecular Oncology, Institute for Medical Research, University of Belgrade, Serbia.,Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| |
Collapse
|
6
|
Sow HS, Ren J, Camps M, Ossendorp F, Ten Dijke P. Combined Inhibition of TGF-β Signaling and the PD-L1 Immune Checkpoint Is Differentially Effective in Tumor Models. Cells 2019; 8:cells8040320. [PMID: 30959852 PMCID: PMC6523576 DOI: 10.3390/cells8040320] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 12/14/2022] Open
Abstract
Antibodies blocking the programmed death-ligand 1 (PD-L1) have shown impressive and durable responses in clinical studies. However, this type of immunotherapy is only effective in a subset of patients and not sufficient for rejection of all tumor types. In this study, we explored in two mouse tumor models whether the antitumor effect could be enhanced by the combined blockade of PD-L1 and transforming growth factor-β (TGF-β), a potent immunosuppressive cytokine. The effect of anti-PD-L1 mouse monoclonal (mAb) and a TGF-β type I receptor small molecule kinase inhibitor (LY364947) was evaluated in the highly immunogenic mouse MC38 colon adenocarcinoma and the poorly immunogenic mouse KPC1 pancreatic tumor model. In the MC38 tumor model, LY364947 monotherapy did not show any antitumor effect, whereas treatment with anti-PD-L1 mAb significantly delayed tumor outgrowth. However, combination therapy showed the strongest therapeutic efficacy, resulting in improved long-term survival compared with anti-PD-L1 mAb monotherapy. This improved survival was associated with an increased influx of CD8+ T cells in the tumor microenvironment. In the KPC1 tumor model, LY364947 did not enhance the antitumor effect of anti-PD-L1 mAb. Despite this, delayed KPC1 tumor outgrowth was observed in the LY364947-treated group and this treatment led to a significant reduction of CD4+ T cells in the tumor microenvironment. Together, our data indicate that an additive anti-tumor response of dual targeting PD-L1 and TGF-β is dependent on the tumor model used, highlighting the importance of selecting appropriate cancer types, using in-depth analysis of the tumor microenvironment, which can benefit from combinatorial immunotherapy regimens.
Collapse
Affiliation(s)
- Heng Sheng Sow
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
| | - Jiang Ren
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
| | - Marcel Camps
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
| |
Collapse
|
7
|
Gurska LM, Ames K, Gritsman K. Signaling Pathways in Leukemic Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1143:1-39. [PMID: 31338813 PMCID: PMC7249489 DOI: 10.1007/978-981-13-7342-8_1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hematopoietic stem cells (HSCs) and leukemic stem cells (LSCs) utilize many of the same signaling pathways for their maintenance and survival. In this review, we will focus on several signaling pathways whose roles have been extensively studied in both HSCs and LSCs. Our main focus will be on the PI3K/AKT/mTOR pathway and several of its regulators and downstream effectors. We will also discuss several other signaling pathways of particular importance in LSCs, including the WNT/β-catenin pathway, the NOTCH pathway, and the TGFβ pathway. For each of these pathways, we will emphasize differences in how these pathways operate in LSCs, compared to their function in HSCs, to highlight opportunities for the specific therapeutic targeting of LSCs. We will also highlight areas of crosstalk between multiple signaling pathways that may affect LSC function.
Collapse
Affiliation(s)
- Lindsay M Gurska
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kristina Ames
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kira Gritsman
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA.
- Department of Medical Oncology, Montefiore Hospital, Bronx, New York, USA.
| |
Collapse
|
8
|
Binder S, Luciano M, Horejs-Hoeck J. The cytokine network in acute myeloid leukemia (AML): A focus on pro- and anti-inflammatory mediators. Cytokine Growth Factor Rev 2018; 43:8-15. [PMID: 30181021 DOI: 10.1016/j.cytogfr.2018.08.004] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 08/22/2018] [Indexed: 12/15/2022]
Abstract
Cytokines exert profound effects on the progression of hematopoietic malignancies such as acute myeloid leukemia (AML). Critical roles of cytokines in the context of inflammation have gained special interest. While pro-inflammatory mediators such as IL-1β, TNF-α and IL-6 tend to increase AML aggressiveness, anti-inflammatory mediators such as TGF-β and IL-10 appear to impede AML progression. Dysregulation of the complex interactions between pro- and anti-inflammatory cytokines in AML may create a pro-tumorigenic microenvironment with effects on leukemic cell proliferation, survival and drug-resistance. This article summarizes current knowledge about the functions of pro- and anti-inflammatory cytokines in AML, their modes of action, and therapeutic interventions with potential to improve clinical outcomes for AML patients.
Collapse
Affiliation(s)
- Stephanie Binder
- University of Salzburg, Department of Biosciences, Hellbrunner Str. 34, Salzburg, Austria
| | - Michela Luciano
- University of Salzburg, Department of Biosciences, Hellbrunner Str. 34, Salzburg, Austria
| | - Jutta Horejs-Hoeck
- University of Salzburg, Department of Biosciences, Hellbrunner Str. 34, Salzburg, Austria.
| |
Collapse
|
9
|
Wong ACH, Rasko JEJ, Wong JJL. We skip to work: alternative splicing in normal and malignant myelopoiesis. Leukemia 2018; 32:1081-1093. [DOI: 10.1038/s41375-018-0021-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/14/2017] [Accepted: 12/22/2017] [Indexed: 12/15/2022]
|