1
|
Li C, Lu W, Zhang H. BTB domain and CNC homolog 2: A master regulator that controls immune response and cancer progression. Biochim Biophys Acta Rev Cancer 2025:189325. [PMID: 40252853 DOI: 10.1016/j.bbcan.2025.189325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
BTB domain and CNC homolog 2 (BACH2) is a transcription repressor from the basic region leucine zipper (bZIP) family. Although BACH2 is predominantly expressed in lymphoid cells, it plays pivotal roles throughout hematological development and differentiation, ranging from the regulation of hematopoietic stem and progenitor cell (HSPC) lineage commitment to the development of both innate and adaptive immune cells. Given its extensive regulation of immunity, it is not surprising that BACH2 has been implicated in cancer, particularly in hematological malignancies. While multiple findings indicate that BACH2 acts primarily as a tumor suppressor, other findings suggest that BACH2, whether within tumor cells or their surrounding microenvironment, may contribute to tumorigenesis and progression, highlighting the complexity of its roles and the diverse networks involved in different contexts. In this review, we provide a comprehensive overview of the evolving roles of BACH2 across various stages of hematopoiesis, with a particular focus on its associations with cancer and its therapeutic potential in a wide range of cancer types.
Collapse
Affiliation(s)
- Chenyang Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, 650118, China
| | - Wei Lu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, 650118, China
| | - Han Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, 650118, China.
| |
Collapse
|
2
|
Sheng Z, Pan Y, Shao L, Bao Y. METTL3 mediates CPB1 expression by regulating transcription factor BACH2 to promote apoptosis and oxidative stress of lens epithelial cells. J Bioenerg Biomembr 2025:10.1007/s10863-025-10054-1. [PMID: 39982642 DOI: 10.1007/s10863-025-10054-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/09/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Cataracts are a significant cause of vision loss, adversely affecting the quality of human life. Numerous studies have reported that lens epithelial cells (LECs) play a crucial role in age-related cataract (ARC). However, the roles of carboxypeptidase B 1 (CPB1) and transcription factor BTB and CNC homologue 2 (BACH2) in the pathogenesis of ARC remain unclear. In this study, we aim to explore the contributions of CPB1 and BACH2 to the development of ARC. METHODS The Gene Expression Omnibus (GEO) was utilized to screen for differentially expressed genes. mRNA and protein levels were assessed using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot analysis. Flow cytometry was conducted to analyze apoptosis. The levels of superoxide dismutase (SOD), glutathione peroxidase (GSH-PX), and malondialdehyde (MDA) were measured using a commercial kit. Dual-luciferase reporter assays and chromatin immunoprecipitation (CHIP) were performed to investigate the interaction between CPB1 and BACH2. The methylation site of BACH2 was analyzed using the RNA-protein binding sites prediction suite and the sequence-based RNA adenosine methylation site predictor suite. Methylated RNA immunoprecipitation (Me-RIP) was employed to detect m6A modification level of BACH2. RESULTS In ARC and H2O2-induced human lens epithelial cells (HLECs), CPB1, BACH2, and METTL3 were found to be up-regulated. Silencing CPB1 reduced apoptosis and MDA levels while enhancing the activities of SOD and GSH-PX in H2O2-induced HLECs. Additionally, CPB1 was shown to bind to BACH2, and knockdown of BACH2 attenuated apoptosis and oxidative stress in H2O2-induced HLECs by targeting CPB1. Notably, METTL3 promoted the BACH2 expression by enhancing CPB1 expression in H2O2-induced HLECs. Finally, silencing METTL3 inhibited apoptosis and oxidative stress in H2O2-induced HLECs by hampering BACH2 expression. CONCLUSIONS METTL3 facilitates apoptosis and oxidative stress in H2O2-induced HLECs by promoting the modification of BACH2 and CPB1 expression.
Collapse
Affiliation(s)
- Zhangxing Sheng
- Department of Ophthalmology, Shaoxing Hospital of Traditional Chinese Medicine, No. 641, Renmin Middle Road, Yuecheng District, Shaoxing, 312000, China.
| | - Yu Pan
- Department of Ophthalmology, Shaoxing Hospital of Traditional Chinese Medicine, No. 641, Renmin Middle Road, Yuecheng District, Shaoxing, 312000, China
| | - Liqin Shao
- Department of Ophthalmology, Affiliated Hospital of Shaoxing Art and Sciences University, Shaoxing, China
| | - Yihui Bao
- Department of Ophthalmology, Shaoxing Hospital of Traditional Chinese Medicine, No. 641, Renmin Middle Road, Yuecheng District, Shaoxing, 312000, China
| |
Collapse
|
3
|
Haque S, Diamond B. Myricetin exposure reduces PC differentiation in vitro in primary human B cells. Mol Med 2025; 31:28. [PMID: 39875809 PMCID: PMC11776280 DOI: 10.1186/s10020-025-01068-x] [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: 09/10/2024] [Accepted: 01/03/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND The process of B cell activation and plasma cell (PC) formation involves morphological, transcriptional, and metabolic changes in the B cell. Blocking or reducing PC differentiation is one approach to treat autoimmune diseases that are characterized by the presence of pathogenic autoantibodies. Recent studies have suggested the potential of myricetin, a natural flavonoid with anti-inflammatory and antioxidant properties, to block or reduce PC differentiation. METHODS Primary human B cells were purified by using a human B cell isolation kit. B cell subsets such as IgG memory B cells, marginal zone B cells (MZ B cells), and naive B cells were isolated by flow cytometry and activated to induce PC differentiation. Quantification of PCs (CD27 + + , CD38 +) was obtained by flow cytometry. The expression of mRNA was measured by qPCR. Ig secretion in culture supernatant was measured by ELISA. RESULTS Myricetin treatment significantly reduced PC differentiation in primary human B cells and all B cell subsets. Myricetin exposure reduced Ig production both IgM and IgG, in culture supernatants at day 5. Myricetin treatment led to augmented BACH2 expression and reduced IRF4, BLIMP1, and XBP1 expression compared to control cultures. CONCLUSION Myricetin treatment reduced PC differentiation and Ig secretion by primary human B cells. Targeting B cells in this way may be a therapeutic approach for some autoimmune diseases.
Collapse
Affiliation(s)
- Shabirul Haque
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, New York, 11030, USA
| | - Betty Diamond
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, New York, 11030, USA.
| |
Collapse
|
4
|
Watanabe-Matsui M, Kadoya S, Segawa K, Shima H, Nakagawa T, Nagasawa Y, Hayashi S, Matsumoto M, Ikeda M, Muto A, Ochiai K, Nguyen LC, Doh-Ura K, Shirouzu M, Nakayama K, Murayama K, Igarashi K. Heme regulates protein interactions and phosphorylation of BACH2 intrinsically disordered region in humoral response. iScience 2025; 28:111529. [PMID: 39758820 PMCID: PMC11699347 DOI: 10.1016/j.isci.2024.111529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 09/03/2023] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Heme is known to bind to the intrinsically disordered region (IDR) to regulate protein function. The binding of heme to the IDR of transcription factor BACH2 promotes plasma cell differentiation, but the molecular basis is unknown. Heme was found to increase BACH2 IDR interaction with TANK-binding kinase 1 (TBK1). TBK1 inactivated BACH2 by phosphorylation of its IDR, whereas BACH2 repressed TBK1 gene expression. BACH2 phosphorylation by TBK1 inhibited its interaction with the co-repressor NCOR1 and promoted plasma cell differentiation. Heme also induced BACH2 binding to ubiquitin E3 ligase adaptor FBXO22, which polyubiquitinated BACH2 only in the presence of heme in vitro. Mutations of some of the TBK1-mediated phosphorylation sites promoted BACH2-FBXO22 interaction, while additional mutations abrogated their interaction, suggesting that TBK1 can both inhibit and promote BACH2-FBXO22 interaction. Therefore, heme regulates phosphorylation of BACH2 IDR by TBK1 and its interaction with NCOR1 and FBXO22, leading to de-repression of BACH2 target genes in humoral immunity.
Collapse
Affiliation(s)
- Miki Watanabe-Matsui
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
- The Japan Society for the Promotion of Science (JSPS), Tokyo, Japan
| | - Shun Kadoya
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kei Segawa
- Pharmaceutical Discovery Research Laboratories, Teijin Pharma Limited, Tokyo, Japan
| | - Hiroki Shima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tadashi Nakagawa
- Division of Cell Proliferation, ART, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Clinical Pharmacology, Sanyo-Onoda City University, Sanyo-Onoda, Japan
| | - Yuko Nagasawa
- Division of Cell Proliferation, ART, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shuichiro Hayashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mariko Ikeda
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Akihiko Muto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kyoko Ochiai
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Long C. Nguyen
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Katsumi Doh-Ura
- Department of Neurochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Keiko Nakayama
- Division of Cell Proliferation, ART, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazutaka Murayama
- Division of Biomedical Measurements and Diagnostics, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
5
|
Rondeau V, Kalogeraki M, Roland L, Nader ZA, Gourhand V, Bonaud A, Lemos J, Khamyath M, Moulin C, Schell B, Delord M, Bidaut G, Lecourt S, Freitas C, Anginot A, Mazure N, McDermott DH, Parietti V, Setterblad N, Dulphy N, Bachelerie F, Aurrand-Lions M, Stockholm D, Lobry C, Murphy PM, Espéli M, Mancini SJ, Balabanian K. CXCR4 signaling determines the fate of hematopoietic multipotent progenitors by stimulating mTOR activity and mitochondrial metabolism. Sci Signal 2024; 17:eadl5100. [PMID: 39471249 PMCID: PMC11733996 DOI: 10.1126/scisignal.adl5100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 05/23/2024] [Accepted: 09/30/2024] [Indexed: 11/01/2024]
Abstract
Both cell-intrinsic and niche-derived, cell-extrinsic cues drive the specification of hematopoietic multipotent progenitors (MPPs) in the bone marrow, which comprise multipotent MPP1 cells and lineage-restricted MPP2, MPP3, and MPP4 subsets. Patients with WHIM syndrome, a rare congenital immunodeficiency caused by mutations that prevent desensitization of the chemokine receptor CXCR4, have an excess of myeloid cells in the bone marrow. Here, we investigated the effects of increased CXCR4 signaling on the localization and fate of MPPs. Knock-in mice bearing a WHIM syndrome-associated CXCR4 mutation (CXCR41013) phenocopied the myeloid skewing of bone marrow in patients. Whereas MPP4 cells in wild-type mice differentiated into lymphoid cells, MPP4s in CXCR41013 knock-in mice differentiated into myeloid cells. This myeloid rewiring of MPP4s in CXCR41013 knock-in mice was associated with enhanced signaling mediated by the kinase mTOR and increased oxidative phosphorylation (OXPHOS). MPP4s also localized further from arterioles in the bone marrow of knock-in mice compared with wild-type mice, suggesting that the loss of extrinsic cues from the perivascular niche may also contribute to their myeloid skewing. Chronic treatment with the CXCR4 antagonist AMD3100 or the mTOR inhibitor rapamycin restored the lymphoid potential of MPP4s in knock-in mice. Thus, CXCR4 desensitization drives the lymphoid potential of MPP4 cells by dampening the mTOR-dependent metabolic changes that promote myeloid differentiation.
Collapse
Affiliation(s)
- Vincent Rondeau
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Maria Kalogeraki
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Lilian Roland
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Zeina Abou Nader
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Vanessa Gourhand
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Amélie Bonaud
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Julia Lemos
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Mélanie Khamyath
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Clémentine Moulin
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Bérénice Schell
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Marc Delord
- Direction à la recherche clinique et à
l’innovation, Centre hospitalier de Versailles, Le Chesnay, France
| | - Ghislain Bidaut
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes,
CRCM, Marseille, France
| | - Séverine Lecourt
- Inserm U1279, Gustave Roussy Cancer Center,
Université Paris Saclay, Villejuif, France
| | - Christelle Freitas
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Adrienne Anginot
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Nathalie Mazure
- Centre Méditerranéen de Médecine
Moléculaire, INSERM U1065, Nice, France
| | - David H. McDermott
- Molecular Signaling Section, Laboratory of Molecular
Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda,
MD, United States
| | - Véronique Parietti
- Université Paris Cité, UMS Saint-Louis INSERM
U53/UAR2030, Paris, France
| | - Niclas Setterblad
- Université Paris Cité, UMS Saint-Louis INSERM
U53/UAR2030, Paris, France
| | - Nicolas Dulphy
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Françoise Bachelerie
- Université Paris-Saclay, INSERM, Inflammation,
Microbiome and Immunosurveillance, Orsay, France
| | - Michel Aurrand-Lions
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
- Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes,
CRCM, Marseille, France
| | - Daniel Stockholm
- PSL Research University, EPHE, Paris, France
- Sorbonne Université, INSERM, Centre de Recherche
Saint-Antoine, CRSA, Paris, France
| | - Camille Lobry
- Université Paris Cité, Institut de
Recherche Saint-Louis, INSERM U944, Paris, France
| | - Philip M. Murphy
- Molecular Signaling Section, Laboratory of Molecular
Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda,
MD, United States
| | - Marion Espéli
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| | | | - Karl Balabanian
- Université Paris Cité, Institut de Recherche
Saint-Louis, INSERM U1160, Paris, France
- OPALE Carnot Institute, The Organization for Partnerships
in Leukemia, Hôpital Saint-Louis, Paris, France
| |
Collapse
|
6
|
Aliazis K, Yenyuwadee S, Phikulsod P, Boussiotis VA. Emergency myelopoiesis in solid cancers. Br J Haematol 2024; 205:798-811. [PMID: 39044285 DOI: 10.1111/bjh.19656] [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: 05/13/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024]
Abstract
Cells of the innate and adaptive immune systems are the progeny of haematopoietic stem and progenitor cells (HSPCs). During steady-state myelopoiesis, HSPC undergo differentiation and proliferation but are called to respond directly and acutely to various signals that lead to emergency myelopoiesis, including bone marrow ablation, infections, and sterile inflammation. There is extensive evidence that many solid tumours have the potential to secrete classical myelopoiesis-promoting growth factors and other products able to mimic emergency haematopoiesis, and to aberrantly re-direct myeloid cell development into immunosuppressive cells with tumour promoting properties. Here, we summarize the current literature regarding the effects of solid cancers on HSPCs function and discuss how these effects might shape antitumour responses via a mechanism initiated at a site distal from the tumour microenvironment.
Collapse
Affiliation(s)
- Konstantinos Aliazis
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Sasitorn Yenyuwadee
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ployploen Phikulsod
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Vassiliki A Boussiotis
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Ma H, Qu J, Pang Z, Luo J, Yan M, Xu W, Zhuang H, Liu L, Qu Q. Super-enhancer omics in stem cell. Mol Cancer 2024; 23:153. [PMID: 39090713 PMCID: PMC11293198 DOI: 10.1186/s12943-024-02066-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/12/2024] [Indexed: 08/04/2024] Open
Abstract
The hallmarks of stem cells, such as proliferation, self-renewal, development, differentiation, and regeneration, are critical to maintain stem cell identity which is sustained by genetic and epigenetic factors. Super-enhancers (SEs), which consist of clusters of active enhancers, play a central role in maintaining stemness hallmarks by specifically transcriptional model. The SE-navigated transcriptional complex, including SEs, non-coding RNAs, master transcriptional factors, Mediators and other co-activators, forms phase-separated condensates, which offers a toggle for directing diverse stem cell fate. With the burgeoning technologies of multiple-omics applied to examine different aspects of SE, we firstly raise the concept of "super-enhancer omics", inextricably linking to Pan-omics. In the review, we discuss the spatiotemporal organization and concepts of SEs, and describe links between SE-navigated transcriptional complex and stem cell features, such as stem cell identity, self-renewal, pluripotency, differentiation and development. We also elucidate the mechanism of stemness and oncogenic SEs modulating cancer stem cells via genomic and epigenetic alterations hijack in cancer stem cell. Additionally, we discuss the potential of targeting components of the SE complex using small molecule compounds, genome editing, and antisense oligonucleotides to treat SE-associated organ dysfunction and diseases, including cancer. This review also provides insights into the future of stem cell research through the paradigm of SEs.
Collapse
Affiliation(s)
- Hongying Ma
- Department of Pharmacy, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, People's Republic of China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Jian Qu
- Department of Pharmacy, the Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, 410011, People's Republic of China
- Hunan key laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, 410219, China
| | - Zicheng Pang
- Department of Pharmacy, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, People's Republic of China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jian Luo
- Department of Pharmacy, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, People's Republic of China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Min Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, People's Republic of China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Weixin Xu
- Department of Pharmacy, the Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, 410011, People's Republic of China
| | - Haihui Zhuang
- Department of Pharmacy, the Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, 410011, People's Republic of China
| | - Linxin Liu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Qiang Qu
- Department of Pharmacy, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, People's Republic of China.
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China.
| |
Collapse
|
8
|
Ochiai K, Shima H, Tamahara T, Sugie N, Funayama R, Nakayama K, Kurosaki T, Igarashi K. Accelerated plasma-cell differentiation in Bach2-deficient mouse B cells is caused by altered IRF4 functions. EMBO J 2024; 43:1947-1964. [PMID: 38605225 PMCID: PMC11099079 DOI: 10.1038/s44318-024-00077-6] [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: 09/13/2023] [Revised: 02/19/2024] [Accepted: 02/24/2024] [Indexed: 04/13/2024] Open
Abstract
Transcription factors BACH2 and IRF4 are both essential for antibody class-switch recombination (CSR) in activated B lymphocytes, while they oppositely regulate the differentiation of plasma cells (PCs). Here, we investigated how BACH2 and IRF4 interact during CSR and plasma-cell differentiation. We found that BACH2 organizes heterochromatin formation of target gene loci in mouse splenic B cells, including targets of IRF4 activation such as Aicda, an inducer of CSR, and Prdm1, a master plasma-cell regulator. Release of these gene loci from heterochromatin in response to B-cell receptor stimulation was coupled to AKT-mTOR pathway activation. In Bach2-deficient B cells, PC genes' activation depended on IRF4 protein accumulation, without an increase in Irf4 mRNA. Mechanistically, a PU.1-IRF4 heterodimer in activated B cells promoted BACH2 function by inducing gene expression of Bach2 and Pten, a negative regulator of AKT signaling. Elevated AKT activity in Bach2-deficient B cells resulted in IRF4 protein accumulation. Thus, BACH2 and IRF4 mutually modulate the activity of each other, and BACH2 inhibits PC differentiation by both the repression of PC genes and the restriction of IRF4 protein accumulation.
Collapse
Affiliation(s)
- Kyoko Ochiai
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai, 980-8575, Japan.
| | - Hiroki Shima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai, 980-8575, Japan
| | - Toru Tamahara
- Division of Community Oral Health Science, Department of Community Medical Supports, Tohoku Medical Megabank Organization, Tohoku University, Seiryo-machi 2-1, Sendai, 980-8573, Japan
| | - Nao Sugie
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai, 980-8575, Japan
| | - Ryo Funayama
- Division of Cell Proliferation, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai, 980-8575, Japan
| | - Keiko Nakayama
- Division of Cell Proliferation, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai, 980-8575, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, Immunology Frontier Research Center, Osaka University, Osaka, 565-0871, Japan
- Laboratory for Lymhocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa, 230-0045, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai, 980-8575, Japan.
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai, 980-8575, Japan.
| |
Collapse
|
9
|
Liu L, Matsumoto M, Watanabe-Matsui M, Nakagawa T, Nagasawa Y, Pang J, Callens BKK, Muto A, Ochiai K, Takekawa H, Alam M, Nishizawa H, Shirouzu M, Shima H, Nakayama K, Igarashi K. TANK Binding Kinase 1 Promotes BACH1 Degradation through Both Phosphorylation-Dependent and -Independent Mechanisms without Relying on Heme and FBXO22. Int J Mol Sci 2024; 25:4141. [PMID: 38673728 PMCID: PMC11050367 DOI: 10.3390/ijms25084141] [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: 02/20/2024] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
BTB and CNC homology 1 (BACH1) represses the expression of genes involved in the metabolism of iron, heme and reactive oxygen species. While BACH1 is rapidly degraded when it is bound to heme, it remains unclear how BACH1 degradation is regulated under other conditions. We found that FBXO22, a ubiquitin ligase previously reported to promote BACH1 degradation, polyubiquitinated BACH1 only in the presence of heme in a highly purified reconstitution assay. In parallel to this regulatory mechanism, TANK binding kinase 1 (TBK1), a protein kinase that activates innate immune response and regulates iron metabolism via ferritinophagy, was found to promote BACH1 degradation when overexpressed in 293T cells. While TBK1 phosphorylated BACH1 at multiple serine and threonine residues, BACH1 degradation was observed with not only the wild-type TBK1 but also catalytically impaired TBK1. The BACH1 degradation in response to catalytically impaired TBK1 was not dependent on FBXO22 but involved both autophagy-lysosome and ubiquitin-proteasome pathways judging from its suppression by using inhibitors of lysosome and proteasome. Chemical inhibition of TBK1 in hepatoma Hepa1 cells showed that TBK1 was not required for the heme-induced BACH1 degradation. Its inhibition in Namalwa B lymphoma cells increased endogenous BACH1 protein. These results suggest that TBK1 promotes BACH1 degradation in parallel to the FBXO22- and heme-dependent pathway, placing BACH1 as a downstream effector of TBK1 in iron metabolism or innate immune response.
Collapse
Affiliation(s)
- Liang Liu
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan (H.T.); (M.A.)
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan (H.T.); (M.A.)
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan
| | - Miki Watanabe-Matsui
- Department of Neurochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan
| | - Tadashi Nakagawa
- Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan; (T.N.); (K.N.)
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda 756-0884, Japan
| | - Yuko Nagasawa
- Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan; (T.N.); (K.N.)
| | - Jingyao Pang
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan (H.T.); (M.A.)
| | - Bert K. K. Callens
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan (H.T.); (M.A.)
- Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 GT Maastricht, The Netherlands
| | - Akihiko Muto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan (H.T.); (M.A.)
| | - Kyoko Ochiai
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan (H.T.); (M.A.)
| | - Hirotaka Takekawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan (H.T.); (M.A.)
| | - Mahabub Alam
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan (H.T.); (M.A.)
- Department of Animal Science and Nutrition, Chattogram Veterinary and Animal Sciences University, Khulshi, Chattogram 4225, Bangladesh
| | - Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan (H.T.); (M.A.)
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama 305-0074, Japan
| | - Hiroki Shima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan (H.T.); (M.A.)
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan
| | - Keiko Nakayama
- Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan; (T.N.); (K.N.)
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan (H.T.); (M.A.)
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan
| |
Collapse
|
10
|
Nagai K, Niihori T, Muto A, Hayashi Y, Abe T, Igarashi K, Aoki Y. Mecom mutation related to radioulnar synostosis with amegakaryocytic thrombocytopenia reduces HSPCs in mice. Blood Adv 2023; 7:5409-5420. [PMID: 37099686 PMCID: PMC10509669 DOI: 10.1182/bloodadvances.2022008462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 04/28/2023] Open
Abstract
Radioulnar synostosis with amegakaryocytic thrombocytopenia (RUSAT) is an inherited bone marrow failure syndrome characterized by the congenital fusion of the forearm bones. RUSAT is largely caused by missense mutations that are clustered in a specific region of the MDS1 and EVI1 complex locus (MECOM). EVI1, a transcript variant encoded by MECOM, is a zinc finger transcription factor involved in hematopoietic stem cell maintenance that induce leukemic transformation when overexpressed. Mice with exonic deletions in Mecom show reduced hematopoietic stem and progenitor cells (HSPCs). However, the pathogenic roles of RUSAT-associated MECOM mutations in vivo have not yet been elucidated. To investigate the impact of the RUSAT-associated MECOM mutation on the phenotype, we generated knockin mice harboring a point mutation (translated into EVI1 p.H752R and MDS1-EVI1 p.H942R), which corresponds to an EVI1 p.H751R and MDS1-EVI1 p.H939R mutation identified in a patient with RUSAT. Homozygous mutant mice died at embryonic day 10.5 to 11.5. Heterozygous mutant mice (Evi1KI/+ mice) grew normally without radioulnar synostosis. Male Evi1KI/+ mice, aged between 5 and 15 weeks, exhibited lower body weight, and those aged ≥16 weeks showed low platelet counts. Flow cytometric analysis of bone marrow cells revealed a decrease in HSPCs in Evi1KI/+ mice between 8 and 12 weeks. Moreover, Evi1KI/+ mice showed delayed leukocyte and platelet recovery after 5-fluorouracil-induced myelosuppression. These findings suggest that Evi1KI/+ mice recapitulate the bone marrow dysfunction in RUSAT, similar to that caused by loss-of-function Mecom alleles.
Collapse
Affiliation(s)
- Koki Nagai
- Department of Medical Genetics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsuya Niihori
- Department of Medical Genetics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akihiko Muto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshikazu Hayashi
- Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Taiki Abe
- Department of Medical Genetics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoko Aoki
- Department of Medical Genetics, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
11
|
Zhu N, Hu L, Hu W, Li Q, Mao H, Wang M, Ke Z, Qi L, Wang J. Comparative Transcriptome Profiling of mRNA and lncRNA of Mouse Spleens Inoculated with the Group ACYW135 Meningococcal Polysaccharide Vaccine. Vaccines (Basel) 2023; 11:1295. [PMID: 37631863 PMCID: PMC10458039 DOI: 10.3390/vaccines11081295] [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: 05/17/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
The Group ACYW135 meningococcal polysaccharide vaccine (MPV-ACYW135) is a classical common vaccine used to prevent Neisseria meningitidis serogroups A, C, Y, and W135, but studies on the vaccine at the transcriptional level are still limited. In the present study, mRNAs and lncRNAs related to immunity were screened from the spleens of mice inoculated with MPV-ACYW135 and compared with the control group to identify differentially expressed mRNAs and lncRNAs in the immune response. The result revealed 34375 lncRNAs and 41321 mRNAs, including 405 differentially expressed (DE) lncRNAs and 52 DE mRNAs between the MPV group and the control group. Results of GO and KEGG enrichment analysis turned out that the main pathways related to the immunity of target genes of those DE mRNAs and DE lncRNAs were largely associated with positive regulation of T cell activation, CD8-positive immunoglobulin production in mucosal tissue, alpha-beta T cell proliferation, negative regulation of CD4-positive, and negative regulation of interleukin-17 production, suggesting that the antigens of MPV-ACYW135 capsular polysaccharide might activate T cell related immune reaction in the vaccine inoculation. In addition, it was noted that Bach2 (BTB and CNC homolog 2), the target gene of lncRNA MSTRG.17645, was involved in the regulation of immune response in MPV-ACYW135 vaccination. This study provided a preliminary catalog of both mRNAs and lncRNAs associated with the proliferation and differentiation of body immune cells, which was worthy of further research to enhance the understanding of the biological immune process regulated by MPV-ACYW135.
Collapse
Affiliation(s)
- Nan Zhu
- School of Biological and Chemical Engineering, NingboTech University, Qianhunan Road 1, Ningbo 315100, China; (N.Z.); (L.H.); (W.H.); (Q.L.); (M.W.); (Z.K.); (L.Q.)
- Aimei Vacin BioPharm (Zhejiang) Co., Ltd., Ningbo 315000, China
| | - Liping Hu
- School of Biological and Chemical Engineering, NingboTech University, Qianhunan Road 1, Ningbo 315100, China; (N.Z.); (L.H.); (W.H.); (Q.L.); (M.W.); (Z.K.); (L.Q.)
- Aimei Vacin BioPharm (Zhejiang) Co., Ltd., Ningbo 315000, China
| | - Wenlong Hu
- School of Biological and Chemical Engineering, NingboTech University, Qianhunan Road 1, Ningbo 315100, China; (N.Z.); (L.H.); (W.H.); (Q.L.); (M.W.); (Z.K.); (L.Q.)
- Aimei Vacin BioPharm (Zhejiang) Co., Ltd., Ningbo 315000, China
| | - Qiang Li
- School of Biological and Chemical Engineering, NingboTech University, Qianhunan Road 1, Ningbo 315100, China; (N.Z.); (L.H.); (W.H.); (Q.L.); (M.W.); (Z.K.); (L.Q.)
- Aimei Vacin BioPharm (Zhejiang) Co., Ltd., Ningbo 315000, China
| | - Haiguang Mao
- School of Biological and Chemical Engineering, NingboTech University, Qianhunan Road 1, Ningbo 315100, China; (N.Z.); (L.H.); (W.H.); (Q.L.); (M.W.); (Z.K.); (L.Q.)
| | - Mengting Wang
- School of Biological and Chemical Engineering, NingboTech University, Qianhunan Road 1, Ningbo 315100, China; (N.Z.); (L.H.); (W.H.); (Q.L.); (M.W.); (Z.K.); (L.Q.)
| | - Zhijian Ke
- School of Biological and Chemical Engineering, NingboTech University, Qianhunan Road 1, Ningbo 315100, China; (N.Z.); (L.H.); (W.H.); (Q.L.); (M.W.); (Z.K.); (L.Q.)
| | - Lili Qi
- School of Biological and Chemical Engineering, NingboTech University, Qianhunan Road 1, Ningbo 315100, China; (N.Z.); (L.H.); (W.H.); (Q.L.); (M.W.); (Z.K.); (L.Q.)
| | - Jinbo Wang
- School of Biological and Chemical Engineering, NingboTech University, Qianhunan Road 1, Ningbo 315100, China; (N.Z.); (L.H.); (W.H.); (Q.L.); (M.W.); (Z.K.); (L.Q.)
| |
Collapse
|
12
|
Lieske A, Agyeman-Duah E, Selich A, Dörpmund N, Talbot SR, Schambach A, Maetzig T. A pro B cell population forms the apex of the leukemic hierarchy in Hoxa9/Meis1-dependent AML. Leukemia 2023; 37:79-90. [PMID: 36517672 PMCID: PMC9883166 DOI: 10.1038/s41375-022-01775-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022]
Abstract
Relapse is a major challenge to therapeutic success in acute myeloid leukemia (AML) and can be partly associated with heterogeneous leukemic stem cell (LSC) properties. In the murine Hoxa9/Meis1-dependent (H9M) AML model, LSC potential lies in three defined immunophenotypes, including Lin-cKit+ progenitor cells (Lin-), Gr1+CD11b+cKit+ myeloid cells, and lymphoid cells (Lym+). Previous reports demonstrated their interconversion and distinct drug sensitivities. In contrast, we here show that H9M AML is hierarchically organized. We, therefore, tracked the developmental potential of LSC phenotypes. This unexpectedly revealed a substantial fraction of Lin- LSCs that failed to regenerate Lym+ LSCs, and that harbored reduced leukemogenic potential. However, Lin- LSCs capable of producing Lym+ LSCs as well as Lym+ LSCs triggered rapid disease development suggestive of their high relapse-driving potential. Transcriptional analyses revealed that B lymphoid master regulators, including Sox4 and Bach2, correlated with Lym+ LSC development and presumably aggressive disease. Lentiviral overexpression of Sox4 and Bach2 induced dedifferentiation of H9M cells towards a lineage-negative state in vitro as the first step of lineage conversion. This work suggests that the potency to initiate a partial B lymphoid primed transcriptional program as present in infant AML correlates with aggressive disease and governs the H9M LSC hierarchy.
Collapse
Affiliation(s)
- Anna Lieske
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Eric Agyeman-Duah
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Anton Selich
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Nicole Dörpmund
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Steven R Talbot
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tobias Maetzig
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
13
|
Nishizawa H, Yamanaka M, Igarashi K. Ferroptosis: regulation by competition between NRF2 and BACH1 and propagation of the death signal. FEBS J 2022; 290:1688-1704. [PMID: 35107212 DOI: 10.1111/febs.16382] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/25/2022] [Accepted: 01/31/2022] [Indexed: 12/15/2022]
Abstract
Ferroptosis is triggered by a chain of intracellular labile iron-dependent peroxidation of cell membrane phospholipids. Ferroptosis is important not only as a cause of ischaemic and neurodegenerative diseases but also as a mechanism of cancer suppression, and a better understanding of its regulatory mechanism is required. It has become clear that ferroptosis is finely controlled by two oxidative stress-responsive transcription factors, NRF2 (NF-E2-related factor 2) and BACH1 (BTB and CNC homology 1). NRF2 and BACH1 inhibit and promote ferroptosis, respectively, by activating or suppressing the expression of genes in the major regulatory pathways of ferroptosis: intracellular labile iron metabolism, the GSH (glutathione) -GPX4 (glutathione peroxidase 4) pathway and the FSP1 (ferroptosis suppressor protein 1)-CoQ (coenzyme Q) pathway. In addition to this, NRF2 and BACH1 control ferroptosis through the regulation of lipid metabolism and cell differentiation. This multifaceted regulation of ferroptosis by NRF2 and BACH1 is considered to have been acquired during the evolution of multicellular organisms, allowing the utilization of ferroptosis for maintaining homeostasis, including cancer suppression. In terms of cell-cell interaction, it has been revealed that ferroptosis has the property of propagating to surrounding cells along with lipid peroxidation. The regulation of ferroptosis by NRF2 and BACH1 and the propagation phenomenon could be used to realize anticancer cell therapy in the future. In this review, these points will be summarized and discussed.
Collapse
Affiliation(s)
- Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mie Yamanaka
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan.,Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
14
|
Igarashi K, Nishizawa H, Matsumoto M. Iron in Cancer Progression: Does BACH1 Promote Metastasis by Altering Iron Homeostasis? Subcell Biochem 2022; 100:67-80. [PMID: 36301491 DOI: 10.1007/978-3-031-07634-3_2] [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] [Indexed: 06/16/2023]
Abstract
The transcription factor BACH1, which is regulated by direct binding of prosthetic group heme, promotes epithelial-mesenchymal transition (EMT) and drives metastasis of diverse types of cancer cells. De-regulated target genes of BACH1 in cancer cells include those for glycolysis, oxidative phosphorylation, epithelial cell adhesion, and mesodermal cell motility. In addition, the canonical target genes of BACH1 include genes for the regulation of iron homeostasis. Importantly, cancer cells are addicted to iron. We summarize known functions of BACH1 in cancer and discuss how BACH1 may affect iron homeostasis in cancer cells to support their progression by increasing mobile iron within cells. The dependency on BACH1 for cancer progression may also confer upon cancer cells susceptibility to iron-dependent cell death ferroptosis. Finally, we discuss that the human transcription factors provide research opportunities for better understanding of cancer cell properties.
Collapse
Affiliation(s)
- Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
15
|
Raczkowski HL, Xu LS, Wang WC, Dekoter RP. The E26 Transformation-Specific Family Transcription Factor Spi-C Is Dynamically Regulated by External Signals in B Cells. Immunohorizons 2022; 6:104-115. [PMID: 38285436 DOI: 10.4049/immunohorizons.2100111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 12/31/2021] [Indexed: 01/30/2024] Open
Abstract
Spi-C is an E26 transformation-specific transcription factor closely related to PU.1 and Spi-B. Spi-C has lineage-instructive functions important in B cell development, Ab-generating responses, and red pulp macrophage generation. This research examined the regulation of Spi-C expression in mouse B cells. To determine the mechanism of Spic regulation, we identified the Spic promoter and upstream regulatory elements. The Spic promoter had unidirectional activity that was reduced by mutation of an NF-κB binding site. Reverse transcription-quantitative PCR analysis revealed that Spic expression was reduced in B cells following treatment with cytokines BAFF + IL-4 + IL-5, anti-IgM Ab, or LPS. Cytochalasin treatment partially prevented downregulation of Spic. Unstimulated B cells upregulated Spic on culture. Spic was repressed by an upstream regulatory region interacting with the heme-binding regulator Bach2. Taken together, these data indicate that Spi-C is dynamically regulated by external signals in B cells and provide insight into the mechanism of regulation.
Collapse
Affiliation(s)
- Hannah L Raczkowski
- Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Division of Genetics and Development, Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Li S Xu
- Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Division of Genetics and Development, Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Wei Cen Wang
- Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Division of Genetics and Development, Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Rodney P Dekoter
- Department of Microbiology & Immunology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Division of Genetics and Development, Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
16
|
Hu RD, Zhang W, Li L, Zuo ZQ, Ma M, Ma JF, Yin TT, Gao CY, Yang SH, Zhao ZB, Li ZJ, Qiao GB, Lian ZX, Qu K. Chromatin accessibility analysis identifies the transcription factor ETV5 as a suppressor of adipose tissue macrophage activation in obesity. Cell Death Dis 2021; 12:1023. [PMID: 34716308 PMCID: PMC8556336 DOI: 10.1038/s41419-021-04308-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 12/25/2022]
Abstract
Activation of adipose tissue macrophages (ATMs) contributes to chronic inflammation and insulin resistance in obesity. However, the transcriptional regulatory machinery involved in ATM activation during the development of obesity is not fully understood. Here, we profiled the chromatin accessibility of blood monocytes and ATMs from obese and lean mice using assay for transposase-accessible chromatin sequencing (ATAC-seq). We found that monocytes and ATMs from obese and lean mice exhibited distinct chromatin accessibility status. There are distinct regulatory elements that are specifically associated with monocyte or ATM activation in obesity. We also discovered several transcription factors that may regulate monocyte and ATM activation in obese mice, specifically a predicted transcription factor named ETS translocation variant 5 (ETV5). The expression of ETV5 was significantly decreased in ATMs from obese mice and its downregulation was mediated by palmitate stimulation. The decrease in ETV5 expression resulted in macrophage activation. Our results also indicate that ETV5 suppresses endoplasmic reticulum (ER) stress and Il6 expression in macrophages. Our work delineates the changes in chromatin accessibility in monocytes and ATMs during obesity, and identifies ETV5 as a critical transcription factor suppressing ATM activation, suggesting its potential use as a therapeutic target in obesity-related chronic inflammation.
Collapse
Affiliation(s)
- Ren-Dong Hu
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Wen Zhang
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui, China
| | - Liang Li
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - Zu-Qi Zuo
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui, China
| | - Min Ma
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jin-Fen Ma
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Ting-Ting Yin
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong, China
| | - Cai-Yue Gao
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Shu-Han Yang
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Zhi-Bin Zhao
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zi-Jun Li
- Guangdong Provincial Institute of Geriatrics, Concord Medical Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Gui-Bin Qiao
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhe-Xiong Lian
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China. .,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China.
| | - Kun Qu
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui, China.
| |
Collapse
|
17
|
SON inhibits megakaryocytic differentiation via repressing RUNX1 and the megakaryocytic gene expression program in acute megakaryoblastic leukemia. Cancer Gene Ther 2021; 28:1000-1015. [PMID: 33247227 PMCID: PMC8155101 DOI: 10.1038/s41417-020-00262-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/07/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023]
Abstract
A high incidence of acute megakaryoblastic leukemia (AMKL) in Down syndrome patients implies that chromosome 21 genes have a pivotal role in AMKL development, but the functional contribution of individual genes remains elusive. Here, we report that SON, a chromosome 21-encoded DNA- and RNA-binding protein, inhibits megakaryocytic differentiation by suppressing RUNX1 and the megakaryocytic gene expression program. As megakaryocytic progenitors differentiate, SON expression is drastically reduced, with mature megakaryocytes having the lowest levels. In contrast, AMKL cells express an aberrantly high level of SON, and knockdown of SON induced the onset of megakaryocytic differentiation in AMKL cell lines. Genome-wide transcriptome analyses revealed that SON knockdown turns on the expression of pro-megakaryocytic genes while reducing erythroid gene expression. Mechanistically, SON represses RUNX1 expression by directly binding to the proximal promoter and two enhancer regions, the known +23 kb enhancer and the novel +139 kb enhancer, at the RUNX1 locus to suppress H3K4 methylation. In addition, SON represses the expression of the AP-1 complex subunits JUN, JUNB, and FOSB which are required for late megakaryocytic gene expression. Our findings define SON as a negative regulator of RUNX1 and megakaryocytic differentiation, implicating SON overexpression in impaired differentiation during AMKL development.
Collapse
|
18
|
IDH1 mutation contributes to myeloid dysplasia in mice by disturbing heme biosynthesis and erythropoiesis. Blood 2021; 137:945-958. [PMID: 33254233 DOI: 10.1182/blood.2020007075] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 11/18/2020] [Indexed: 12/22/2022] Open
Abstract
Isocitrate dehydrogenase (IDH) mutations are common genetic alterations in myeloid disorders, including acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Epigenetic changes, including abnormal histone and DNA methylation, have been implicated in the pathogenic build-up of hematopoietic progenitors, but it is still unclear whether and how IDH mutations themselves affect hematopoiesis. Here, we show that IDH1-mutant mice develop myeloid dysplasia in that these animals exhibit anemia, ineffective erythropoiesis, and increased immature progenitors and erythroblasts. In erythroid cells of these mice, D-2-hydroxyglutarate, an aberrant metabolite produced by the mutant IDH1 enzyme, inhibits oxoglutarate dehydrogenase activity and diminishes succinyl-coenzyme A (CoA) production. This succinyl-CoA deficiency attenuates heme biosynthesis in IDH1-mutant hematopoietic cells, thus blocking erythroid differentiation at the late erythroblast stage and the erythroid commitment of hematopoietic stem cells, while the exogenous succinyl-CoA or 5-ALA rescues erythropoiesis in IDH1-mutant erythroid cells. Heme deficiency also impairs heme oxygenase-1 expression, which reduces levels of important heme catabolites such as biliverdin and bilirubin. These deficits result in accumulation of excessive reactive oxygen species that induce the cell death of IDH1-mutant erythroid cells. Our results clearly show the essential role of IDH1 in normal erythropoiesis and describe how its mutation leads to myeloid disorders. These data thus have important implications for the devising of new treatments for IDH-mutant tumors.
Collapse
|
19
|
Ugel S, Canè S, De Sanctis F, Bronte V. Monocytes in the Tumor Microenvironment. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 16:93-122. [PMID: 33497262 DOI: 10.1146/annurev-pathmechdis-012418-013058] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Immunotherapy has revolutionized cancer treatment over the past decade. Nonetheless, prolonged survival is limited to relatively few patients. Cancers enforce a multifaceted immune-suppressive network whose nature is progressively shaped by systemic and local cues during tumor development. Monocytes bridge innate and adaptive immune responses and can affect the tumor microenvironment through various mechanisms that induce immune tolerance, angiogenesis, and increased dissemination of tumor cells. Yet monocytes can also give rise to antitumor effectors and activate antigen-presenting cells. This yin-yang activity relies on the plasticity of monocytes in response to environmental stimuli. In this review, we summarize current knowledge of the ontogeny, heterogeneity, and functions of monocytes and monocyte-derived cells in cancer, pinpointing the main pathways that are important for modeling the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Stefano Ugel
- Section of Immunology, Department of Medicine, University of Verona, Verona 37134, Italy;
| | - Stefania Canè
- Section of Immunology, Department of Medicine, University of Verona, Verona 37134, Italy;
| | - Francesco De Sanctis
- Section of Immunology, Department of Medicine, University of Verona, Verona 37134, Italy;
| | - Vincenzo Bronte
- Section of Immunology, Department of Medicine, University of Verona, Verona 37134, Italy;
| |
Collapse
|
20
|
Jung S, Appleton E, Ali M, Church GM, Del Sol A. A computer-guided design tool to increase the efficiency of cellular conversions. Nat Commun 2021; 12:1659. [PMID: 33712564 PMCID: PMC7954801 DOI: 10.1038/s41467-021-21801-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Human cell conversion technology has become an important tool for devising new cell transplantation therapies, generating disease models and testing gene therapies. However, while transcription factor over-expression-based methods have shown great promise in generating cell types in vitro, they often endure low conversion efficiency. In this context, great effort has been devoted to increasing the efficiency of current protocols and the development of computational approaches can be of great help in this endeavor. Here we introduce a computer-guided design tool that combines a computational framework for prioritizing more efficient combinations of instructive factors (IFs) of cellular conversions, called IRENE, with a transposon-based genomic integration system for efficient delivery. Particularly, IRENE relies on a stochastic gene regulatory network model that systematically prioritizes more efficient IFs by maximizing the agreement of the transcriptional and epigenetic landscapes between the converted and target cells. Our predictions substantially increased the efficiency of two established iPSC-differentiation protocols (natural killer cells and melanocytes) and established the first protocol for iPSC-derived mammary epithelial cells with high efficiency.
Collapse
Affiliation(s)
- Sascha Jung
- Computational Biology Group, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Bizkaia Technology Park, Derio, Spain
| | - Evan Appleton
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Muhammad Ali
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Maastricht University School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| | - George M Church
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- GC Therapeutics, Inc, Cambridge, MA, USA
| | - Antonio Del Sol
- Computational Biology Group, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Bizkaia Technology Park, Derio, Spain.
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia.
| |
Collapse
|
21
|
Zhang H, Zhang R, Zheng X, Sun M, Fan J, Fang C, Tian X, Zheng H. BACH2-mediated FOS confers cytarabine resistance via stromal microenvironment alterations in pediatric ALL. Cancer Sci 2021; 112:1235-1250. [PMID: 33393145 PMCID: PMC7935781 DOI: 10.1111/cas.14792] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/30/2020] [Accepted: 12/30/2020] [Indexed: 12/24/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is an aggressive hematological cancer that mainly affects children. Relapse and chemoresistance result in treatment failure, underlining the need for improved therapies. BTB and CNC homology 2 (BACH2) is a lymphoid-specific transcription repressor recognized as a tumor suppressor in lymphomas, but little is known about its function and regulatory network in pediatric ALL (p-ALL). Herein, we found aberrant BACH2 expression at new diagnosis not only facilitated risk stratification of p-ALL but also served as a sensitive predictor of early treatment response and clinical outcome. Silencing BACH2 in ALL cells increased cell proliferation and accelerated cell cycle progression. BACH2 blockade also promoted cell adhesion to bone marrow stromal cells and conferred cytarabine (Ara-C)-resistant properties to leukemia cells by altering stromal microenvironment. Strikingly, we identified FOS, a transcriptional activator competing with BACH2, as a novel downstream target repressed by BACH2. Blocking FOS by chemical compounds enhanced the effect of Ara-C treatment in both primary p-ALL cells and pre-B-ALL-driven leukemia xenografts and prolonged the survival of tumor-bearing mice. These data highlight an interconnected network of BACH2-FOS, disruption of which could render current chemotherapies more effective and offer a promising therapeutic strategy to overcome Ara-C resistance in p-ALL.
Collapse
Affiliation(s)
- Han Zhang
- Institute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
| | - Ruidong Zhang
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics (Capital Medical University)Key Laboratory of Major Diseases in ChildrenMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalNational Center for Children’s HealthCapital Medical UniversityBeijingChina
| | - Xueling Zheng
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics (Capital Medical University)Key Laboratory of Major Diseases in ChildrenMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalNational Center for Children’s HealthCapital Medical UniversityBeijingChina
| | - Ming Sun
- Institute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
| | - Jia Fan
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics (Capital Medical University)Key Laboratory of Major Diseases in ChildrenMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalNational Center for Children’s HealthCapital Medical UniversityBeijingChina
| | - Chunlian Fang
- Department of Hematology and OncologyKunming Children’s Hospital (Children’s Hospital of Kunming Medical University, Yunnan Children’s Medical Center)KunmingChina
| | - Xin Tian
- Department of Hematology and OncologyKunming Children’s Hospital (Children’s Hospital of Kunming Medical University, Yunnan Children’s Medical Center)KunmingChina
| | - Huyong Zheng
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics (Capital Medical University)Key Laboratory of Major Diseases in ChildrenMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalNational Center for Children’s HealthCapital Medical UniversityBeijingChina
| |
Collapse
|
22
|
C/EBPα induces Ebf1 gene expression in common lymphoid progenitors. PLoS One 2020; 15:e0244161. [PMID: 33332417 PMCID: PMC7746190 DOI: 10.1371/journal.pone.0244161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 12/03/2020] [Indexed: 11/19/2022] Open
Abstract
C/EBPα is required for formation of granulocyte-monocyte progenitors (GMP) and also participates in B lymphopoiesis. The common lymphoid progenitor (CLP) and preproB populations but not proB cells express Cebpa, and pan-hematopoietic deletion of the +37 kb Cebpa enhancer using Mx1-Cre leads not only to reduced GMP but also to 2-fold reduced marrow preproB and >15-fold reduced proB and preB cells. We now show that IL7Rα-Cre-mediated deletion of the +37 kb Cebpa enhancer, which occurs in 89% of Ly6D+ and 65% of upstream Ly6D- CLP, leads to a 2-fold reduction of both preproB and proB cells, and a 3-fold reduction in preB cells, with no impact on GMP numbers. These data support a direct role for C/EBPα during B lineage development, with reduced enhancer deletion in Ly6D- CLP mediated by IL7Rα-Cre diminishing the effect on B lymphopoiesis compared to that seen with Mx1-Cre. Amongst mRNAs encoding key transcriptional regulators that initiate B lymphoid specification (PU.1, E2A, IKAROS, EBF1, FOXO1, and BACH2), only Ebf1 levels are altered in CLP upon Mx1-Cre-mediated Cebpa enhancer deletion, with Ebf1 reduced ~40-fold in Flt3+Sca-1intc-kitintIL7Rα+ CLP. In addition, Cebpa and Ebf1 RNAs were 4- and 14-fold higher in hCD4+ versus hCD4- CLP from Cebpa-hCD4 transgenic mice. Histone modification ChIP-Seq data for CLP indicate the presence of active, intronic Ebf1 enhancers located 270 and 280 kb upstream of the transcription start sites. We identified a cis element in this region that strongly binds C/EBPα using the electrophoretic mobility shift assay. Mutation of this C/EBPα-binding site in an Ebf1 enhancer-TK-luciferase reporter leads to a 4-fold reduction in C/EBPα-mediated trans-activation. These findings support a model of B lymphopoiesis in which induction of Ebf1 by C/EBPα in a subset of CLP contributes to initiation of B lymphopoiesis.
Collapse
|
23
|
Giroux NS, Ding S, McClain MT, Burke TW, Petzold E, Chung HA, Palomino GR, Wang E, Xi R, Bose S, Rotstein T, Nicholson BP, Chen T, Henao R, Sempowski GD, Denny TN, Ko ER, Ginsburg GS, Kraft BD, Tsalik EL, Woods CW, Shen X. Chromatin remodeling in peripheral blood cells reflects COVID-19 symptom severity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.12.04.412155. [PMID: 33300002 PMCID: PMC7724678 DOI: 10.1101/2020.12.04.412155] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SARS-CoV-2 infection triggers highly variable host responses and causes varying degrees of illness in humans. We sought to harness the peripheral blood mononuclear cell (PBMC) response over the course of illness to provide insight into COVID-19 physiology. We analyzed PBMCs from subjects with variable symptom severity at different stages of clinical illness before and after IgG seroconversion to SARS-CoV-2. Prior to seroconversion, PBMC transcriptomes did not distinguish symptom severity. In contrast, changes in chromatin accessibility were associated with symptom severity. Furthermore, single-cell analyses revealed evolution of the chromatin accessibility landscape and transcription factor motif occupancy for individual PBMC cell types. The most extensive remodeling occurred in CD14+ monocytes where sub-populations with distinct chromatin accessibility profiles were associated with disease severity. Our findings indicate that pre-seroconversion chromatin remodeling in certain innate immune populations is associated with divergence in symptom severity, and the identified transcription factors, regulatory elements, and downstream pathways provide potential prognostic markers for COVID-19 subjects.
Collapse
Affiliation(s)
- Nicholas S. Giroux
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Shengli Ding
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Micah T. McClain
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA
- Durham Veterans Affairs Health Care System, Durham, NC, USA
- Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA
| | - Thomas W. Burke
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA
| | - Elizabeth Petzold
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA
| | - Hong A. Chung
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Grecia R. Palomino
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Ergang Wang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Rui Xi
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Shree Bose
- Department of Pharmacology and Cancer Biology, School of Medicine, Duke University, Durham, NC, USA
| | - Tomer Rotstein
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | | | - Tianyi Chen
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA
| | - Ricardo Henao
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA
| | - Gregory D. Sempowski
- Duke Human Vaccine Institute and Department of Medicine, School of Medicine, Duke University, Durham, NC, USA
| | - Thomas N. Denny
- Duke Human Vaccine Institute and Department of Medicine, School of Medicine, Duke University, Durham, NC, USA
| | - Emily R. Ko
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA
| | - Geoffrey S. Ginsburg
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA
| | - Bryan D. Kraft
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA
- Durham Veterans Affairs Health Care System, Durham, NC, USA
| | - Ephraim L. Tsalik
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA
- Durham Veterans Affairs Health Care System, Durham, NC, USA
- Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA
| | - Christopher W. Woods
- Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA
- Durham Veterans Affairs Health Care System, Durham, NC, USA
- Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
24
|
Mola S, Foisy S, Boucher G, Major F, Beauchamp C, Karaky M, Goyette P, Lesage S, Rioux JD. A transcriptome-based approach to identify functional modules within and across primary human immune cells. PLoS One 2020; 15:e0233543. [PMID: 32469933 PMCID: PMC7259617 DOI: 10.1371/journal.pone.0233543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 05/07/2020] [Indexed: 11/20/2022] Open
Abstract
Genome-wide transcriptomic analyses have provided valuable insight into fundamental biology and disease pathophysiology. Many studies have taken advantage of the correlation in the expression patterns of the transcriptome to infer a potential biologic function of uncharacterized genes, and multiple groups have examined the relationship between co-expression, co-regulation, and gene function on a broader scale. Given the unique characteristics of immune cells circulating in the blood, we were interested in determining whether it was possible to identify functional co-expression modules in human immune cells. Specifically, we sequenced the transcriptome of nine immune cell types from peripheral blood cells of healthy donors and, using a combination of global and targeted analyses of genes within co-expression modules, we were able to determine functions for these modules that were cell lineage-specific or shared among multiple cell lineages. In addition, our analyses identified transcription factors likely important for immune cell lineage commitment and/or maintenance.
Collapse
Affiliation(s)
- Saraï Mola
- Centre de recherche, Institut de cardiologie de Montréal, Montréal, Québec, Canada
| | - Sylvain Foisy
- Centre de recherche, Institut de cardiologie de Montréal, Montréal, Québec, Canada
| | - Gabrielle Boucher
- Centre de recherche, Institut de cardiologie de Montréal, Montréal, Québec, Canada
| | - François Major
- Unité de recherche en ingénierie des ARN, Institut de recherche en immunologie et en cancérologie, Montréal, Québec, Canada
- Département d’informatique et de recherche opérationnelle, Université de Montréal, Montréal, Québec, Canada
- Département de biochimie et médecine moléculaire, Université de Montréal, Montréal, Québec, Canada
| | - Claudine Beauchamp
- Centre de recherche, Institut de cardiologie de Montréal, Montréal, Québec, Canada
| | - Mohamad Karaky
- Centre de recherche, Institut de cardiologie de Montréal, Montréal, Québec, Canada
| | - Philippe Goyette
- Centre de recherche, Institut de cardiologie de Montréal, Montréal, Québec, Canada
| | - Sylvie Lesage
- Centre de recherche, Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Québec, Canada
| | - John D. Rioux
- Centre de recherche, Institut de cardiologie de Montréal, Montréal, Québec, Canada
- Département de biochimie et médecine moléculaire, Université de Montréal, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
25
|
Laramée AS, Raczkowski H, Shao P, Batista C, Shukla D, Xu L, Haeryfar SMM, Tesfagiorgis Y, Kerfoot S, DeKoter R. Opposing Roles for the Related ETS-Family Transcription Factors Spi-B and Spi-C in Regulating B Cell Differentiation and Function. Front Immunol 2020; 11:841. [PMID: 32457757 PMCID: PMC7225353 DOI: 10.3389/fimmu.2020.00841] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
Generation of specific antibodies during an immune response to infection or vaccination depends on the ability to rapidly and accurately select clones of antibody-secreting B lymphocytes for expansion. Antigen-specific B cell clones undergo the cell fate decision to differentiate into antibody-secreting plasma cells, memory B cells, or germinal center B cells. The E26-transformation-specific (ETS) transcription factors Spi-B and Spi-C are important regulators of B cell development and function. Spi-B is expressed throughout B cell development and is downregulated upon plasma cell differentiation. Spi-C is highly related to Spi-B and has similar DNA-binding specificity. Heterozygosity for Spic rescues B cell development and B cell proliferation defects observed in Spi-B knockout mice. In this study, we show that heterozygosity for Spic rescued defective IgG1 secondary antibody responses in Spib–/– mice. Plasma cell differentiation was accelerated in Spib–/– B cells. Gene expression, ChIP-seq, and reporter gene analysis showed that Spi-B and Spi-C differentially regulated Bach2, encoding a key regulator of plasma cell and memory B cell differentiation. These results suggest that Spi-B and Spi-C oppose the function of one another to regulate B cell differentiation and function.
Collapse
Affiliation(s)
- Anne-Sophie Laramée
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, ON, Canada
| | - Hannah Raczkowski
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, ON, Canada
| | - Peng Shao
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, ON, Canada
| | - Carolina Batista
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, ON, Canada
| | - Devanshi Shukla
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Li Xu
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, ON, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, ON, Canada
| | - Yodit Tesfagiorgis
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Steven Kerfoot
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Rodney DeKoter
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, ON, Canada
| |
Collapse
|
26
|
Peter IS. The function of architecture and logic in developmental gene regulatory networks. Curr Top Dev Biol 2020; 139:267-295. [PMID: 32450963 DOI: 10.1016/bs.ctdb.2020.04.001] [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] [Indexed: 12/15/2022]
Abstract
An important contribution of systems biology is the insight that biological systems depend on the function of molecular interactions and not just on individual molecules. System level mechanisms are particularly important in the development of animals and plants which depends not just on transcription factors and signaling molecules, but also on regulatory circuits and gene regulatory networks (GRNs). However, since GRNs consist of transcription factors, it can be challenging to assess the function of regulatory circuits independently of the function of regulatory factors. The comparison of different GRNs offers a way to do so and leads to several observations. First, similar regulatory circuits operate in various developmental contexts and in different species, and frequently, these circuits are associated with similar developmental functions. Second, given regulatory circuits are often used at particular positions within the GRN hierarchy. Third, in some GRNs, regulatory circuits are organized in a particular order in respect to each other. And fourth, the evolution of GRNs occurs not just by co-option of regulatory genes but also by rewiring of regulatory linkages between conserved regulatory genes, indicating that the organization of interactions is important. Thus, even though in most instances the function of regulatory circuits remains to be discovered, it becomes evident that the architecture and logic of GRNs are functionally important for the control of genome activity and for the specification of the body plan.
Collapse
Affiliation(s)
- Isabelle S Peter
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States.
| |
Collapse
|
27
|
Coactivation of NF-κB and Notch signaling is sufficient to induce B-cell transformation and enables B-myeloid conversion. Blood 2020; 135:108-120. [PMID: 31697816 DOI: 10.1182/blood.2019001438] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 10/06/2019] [Indexed: 12/12/2022] Open
Abstract
NF-κB and Notch signaling can be simultaneously activated in a variety of B-cell lymphomas. Patients with B-cell lymphoma occasionally develop clonally related myeloid tumors with poor prognosis. Whether concurrent activation of both pathways is sufficient to induce B-cell transformation and whether the signaling initiates B-myeloid conversion in a pathological context are largely unknown. Here, we provide genetic evidence that concurrent activation of NF-κB and Notch signaling in committed B cells is sufficient to induce B-cell lymphomatous transformation and primes common progenitor cells to convert to myeloid lineage through dedifferentiation, not transdifferentiation. Intriguingly, the converted myeloid cells can further transform, albeit at low frequency, into myeloid leukemia. Mechanistically, coactivation of NF-κB and Notch signaling endows committed B cells with the ability to self renew. Downregulation of BACH2, a lymphoma and myeloid gene suppressor, but not upregulation of CEBPα and/or downregulation of B-cell transcription factors, is an early event in both B-cell transformation and myeloid conversion. Interestingly, a DNA hypomethylating drug not only effectively eliminated the converted myeloid leukemia cells, but also restored the expression of green fluorescent protein, which had been lost in converted myeloid leukemia cells. Collectively, our results suggest that targeting NF-κB and Notch signaling will not only improve lymphoma treatment, but also prevent the lymphoma-to-myeloid tumor conversion. Importantly, DNA hypomethylating drugs might efficiently treat these converted myeloid neoplasms.
Collapse
|
28
|
Mármol-Sánchez E, Ramayo-Caldas Y, Quintanilla R, Cardoso TF, González-Prendes R, Tibau J, Amills M. Co-expression network analysis predicts a key role of microRNAs in the adaptation of the porcine skeletal muscle to nutrient supply. J Anim Sci Biotechnol 2020; 11:10. [PMID: 31969983 PMCID: PMC6966835 DOI: 10.1186/s40104-019-0412-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 12/04/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The role of non-coding RNAs in the porcine muscle metabolism is poorly understood, with few studies investigating their expression patterns in response to nutrient supply. Therefore, we aimed to investigate the changes in microRNAs (miRNAs), long intergenic non-coding RNAs (lincRNAs) and mRNAs muscle expression before and after food intake. RESULTS We measured the miRNA, lincRNA and mRNA expression levels in the gluteus medius muscle of 12 gilts in a fasting condition (AL-T0) and 24 gilts fed ad libitum during either 5 h. (AL-T1, N = 12) or 7 h. (AL-T2, N = 12) prior to slaughter. The small RNA fraction was extracted from muscle samples retrieved from the 36 gilts and sequenced, whereas lincRNA and mRNA expression data were already available. In terms of mean and variance, the expression profiles of miRNAs and lincRNAs in the porcine muscle were quite different than those of mRNAs. Food intake induced the differential expression of 149 (AL-T0/AL-T1) and 435 (AL-T0/AL-T2) mRNAs, 6 (AL-T0/AL-T1) and 28 (AL-T0/AL-T2) miRNAs and none lincRNAs, while the number of differentially dispersed genes was much lower. Among the set of differentially expressed miRNAs, we identified ssc-miR-148a-3p, ssc-miR-22-3p and ssc-miR-1, which play key roles in the regulation of glucose and lipid metabolism. Besides, co-expression network analyses revealed several miRNAs that putatively interact with mRNAs playing key metabolic roles and that also showed differential expression before and after feeding. One case example was represented by seven miRNAs (ssc-miR-148a-3p, ssc-miR-151-3p, ssc-miR-30a-3p, ssc-miR-30e-3p, ssc-miR-421-5p, ssc-miR-493-5p and ssc-miR-503) which putatively interact with the PDK4 mRNA, one of the master regulators of glucose utilization and fatty acid oxidation. CONCLUSIONS As a whole, our results evidence that microRNAs are likely to play an important role in the porcine skeletal muscle metabolic adaptation to nutrient availability.
Collapse
Affiliation(s)
- Emilio Mármol-Sánchez
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Yuliaxis Ramayo-Caldas
- Animal Breeding and Genetics Program, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain
| | - Raquel Quintanilla
- Animal Breeding and Genetics Program, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain
| | - Tainã Figueiredo Cardoso
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Present address: Embrapa Pecuária Sudeste, Empresa Brasileira de Pesquisa Agropecuária (EMBRAPA), São Carlos, SP 13560-970 Brazil
| | - Rayner González-Prendes
- Department of Animal Science, Universitat de Lleida - Agrotecnio Center, 25198 Lleida, Spain
| | - Joan Tibau
- Animal Breeding and Genetics Program, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain
| | - Marcel Amills
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| |
Collapse
|
29
|
Jang E, Kim UK, Jang K, Song YS, Cha JY, Yi H, Youn J. Bach2 deficiency leads autoreactive B cells to produce IgG autoantibodies and induce lupus through a T cell-dependent extrafollicular pathway. Exp Mol Med 2019; 51:1-13. [PMID: 31819031 PMCID: PMC6901549 DOI: 10.1038/s12276-019-0352-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/08/2019] [Accepted: 11/04/2019] [Indexed: 12/12/2022] Open
Abstract
Class-switched IgG autoantibodies but not unswitched IgM autoantibodies play a crucial role in the development of systemic lupus erythematosus (SLE). Bach2 is known to be essential for class switch recombination of Ig genes, but recent genomic and clinical studies have suggested an association of Bach2 deficiency with SLE. This study was undertaken to examine the mechanism by which Bach2 regulates the development of SLE. Despite defects in Ig class switch recombination and germinal center formation when actively immunized, Bach2−/− mice spontaneously accumulated IgG autoantibody-secreting cells without germinal center reactions in a regulatory T cell-independent manner, and this phenomenon was accompanied by manifestations akin to SLE. Transcriptome analyses revealed that Bach2 regulated the expression of genes related to germinal center formation and SLE pathogenesis in B cells. B cell-specific deletion of Bach2 was sufficient to impair the development of germinal center B cells but insufficient to promote the production of IgG autoantibodies. Bach2 deficiency caused CD4+ T cells to overexpress Icos and differentiate into extrafollicular helper T cells in a cell-autonomous manner. These findings suggest that Bach2-deficient autoreactive B cells preferentially react at extrafollicular sites to give rise to IgG class-switched pathogenic plasma cells and that this effect requires the help of Bach2-Icoshi helper T cells. Thus, the cell-autonomous roles of Bach2 in B cells and in their cognate CD4+ T cells are required to maintain self-tolerance against SLE. Bach2, a protein that regulates gene expression, is required in the B cells and T cells of the immune system to protect against autoimmune disease. Bach2 deficiency has previously been associated with systemic lupus erythematosus (SLE), but the mechanisms through which it contributes to the development of an immune response against healthy tissue in many parts of the body were unclear. Jeehee Youn at Hanyang University in Seoul, South Korea, and colleagues showed that B cells from mice lacking Bach2 produce self-reactive antibodies and express SLE-related genes. Furthermore, when they specifically deleted Bach2 in T cells, they found that it triggered differentiation into a type of T cell which promoted the maturation of self-reactive B cells. The authors conclude that Bach2 activity in both B cells and T cells is key to maintaining immune self-tolerance.
Collapse
Affiliation(s)
- Eunkyeong Jang
- Laboratory of Autoimmunology, Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul, 04763, Korea
| | - Un Kyo Kim
- Laboratory of Autoimmunology, Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul, 04763, Korea
| | - Kiseok Jang
- Department of Pathology, College of Medicine, Hanyang University, Seoul, 04763, Korea
| | - Young Soo Song
- Department of Pathology, College of Medicine, Hanyang University, Seoul, 04763, Korea
| | - Ji-Young Cha
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21936, Korea
| | - Hansol Yi
- Laboratory of Autoimmunology, Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul, 04763, Korea
| | - Jeehee Youn
- Laboratory of Autoimmunology, Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul, 04763, Korea.
| |
Collapse
|
30
|
Kato H, Igarashi K. To be red or white: lineage commitment and maintenance of the hematopoietic system by the "inner myeloid". Haematologica 2019; 104:1919-1927. [PMID: 31515352 PMCID: PMC6886412 DOI: 10.3324/haematol.2019.216861] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/10/2019] [Indexed: 12/21/2022] Open
Abstract
Differentiation of hematopoietic stem and progenitor cells is tightly regulated depending on environmental changes in order to maintain homeostasis. Transcription factors direct the development of hematopoietic cells, such as GATA-1 for erythropoiesis and PU.1 for myelopoiesis. However, recent findings obtained from single-cell analyses raise the question of whether these transcription factors are "initiators" or just "executors" of differentiation, leaving the initiation of hematopoietic stem and progenitor cell differentiation (i.e. lineage commitment) unclear. While a stochastic process is likely involved in commitment, it cannot fully explain the homeostasis of hematopoiesis nor "on-demand" hematopoiesis in response to environmental changes. Transcription factors BACH1 and BACH2 may regulate both commitment and on-demand hematopoiesis because they control erythroid-myeloid and lymphoid-myeloid differentiation by repressing the myeloid program, and their activities are repressed in response to infectious and inflammatory conditions. We summarize possible mechanisms of lineage commitment of hematopoietic stem and progenitor cells suggested by recent findings and discuss the erythroid and lymphoid commitment of hematopoietic stem and progenitor cells, focusing on the gene regulatory network composed of genes encoding key transcription factors. Surprising similarity exists between commitment to erythroid and lymphoid lineages, including repression of the myeloid program by BACH factors. The suggested gene regulatory network of BACH factors sheds light on the myeloid-based model of hematopoiesis. This model will help to understand the tuning of hematopoiesis in higher eukaryotes in the steady-state condition as well as in emergency conditions, the evolutional history of the system, aging and hematopoietic disorders.
Collapse
Affiliation(s)
- Hiroki Kato
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Present address, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
31
|
Chavakis T, Mitroulis I, Hajishengallis G. Hematopoietic progenitor cells as integrative hubs for adaptation to and fine-tuning of inflammation. Nat Immunol 2019; 20:802-811. [PMID: 31213716 DOI: 10.1038/s41590-019-0402-5] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 04/12/2019] [Indexed: 12/17/2022]
Abstract
Recent advances have highlighted the ability of hematopoietic stem and progenitor cells in the bone marrow to sense peripheral inflammation or infection and adapt through increased proliferation and skewing toward the myeloid lineage. Such adaptations can meet the increased demand for innate immune cells and can be beneficial in response to infection or myeloablation. However, the inflammation-induced adaptation of hematopoietic and myeloid progenitor cells toward enhanced myelopoiesis might also perpetuate inflammation in chronic inflammatory or cardio-metabolic diseases by generating a feed-forward loop between inflammation-adapted hematopoietic progenitor cells and the inflammatory disorder. Sustained adaptive responses of progenitor cells in the bone marrow can also contribute to trained immunity, a non-specific memory of earlier encounters that in turn facilitates the heightened response of these cells, as well as that of their progeny, to future challenges. Here we discuss the mechanisms that govern the adaptation of hematopoietic progenitor cells to inflammation and its sequelae in the pathogenesis of human disease.
Collapse
Affiliation(s)
- Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany.
| | - Ioannis Mitroulis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany.,National Center for Tumor Diseases, Partner Site Dresden, of the German Cancer Research Center, Heidelberg and of the Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, and of the Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,Department of Haematology and Laboratory of Molecular Hematology, Democritus University of Thrace, Alexandroupolis, Greece
| | - George Hajishengallis
- University of Pennsylvania, Penn Dental Medicine, Department of Microbiology, Philadelphia, PA, USA
| |
Collapse
|
32
|
Kurotaki D, Nakabayashi J, Nishiyama A, Sasaki H, Kawase W, Kaneko N, Ochiai K, Igarashi K, Ozato K, Suzuki Y, Tamura T. Transcription Factor IRF8 Governs Enhancer Landscape Dynamics in Mononuclear Phagocyte Progenitors. Cell Rep 2019. [PMID: 29514092 DOI: 10.1016/j.celrep.2018.02.048] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Monocytes and dendritic cells (DCs), mononuclear phagocytes essential for immune responses, develop from hematopoietic stem cells via monocyte-DC progenitors (MDPs). The molecular basis of their development remains unclear. Because promoter-distal enhancers are key to cell fate decisions, we analyzed enhancer landscapes during mononuclear phagocyte development in vivo. Monocyte- and DC-specific enhancers were gradually established at progenitor stages before the expression of associated genes. Of the transcription factors predicted to bind to these enhancers, IRF8, essential for monocyte and DC development, was found to be required for the establishment of these enhancers, particularly those common to both monocyte and DC lineages. Although Irf8-/- mononuclear phagocyte progenitors, including MDPs, displayed grossly normal gene expression patterns, their enhancer landscapes resembled that of an upstream progenitor population. Our results illustrate the dynamic process by which key transcription factors regulate enhancer formation and, therefore, direct future gene expression to achieve mononuclear phagocyte development.
Collapse
Affiliation(s)
- Daisuke Kurotaki
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Jun Nakabayashi
- Advanced Medical Research Center, Yokohama City University, Yokohama 236-0004, Japan
| | - Akira Nishiyama
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Haruka Sasaki
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Wataru Kawase
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Naofumi Kaneko
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Kyoko Ochiai
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Keiko Ozato
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, University of Tokyo, Chiba 277-8562, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan; Advanced Medical Research Center, Yokohama City University, Yokohama 236-0004, Japan.
| |
Collapse
|
33
|
Infection perturbs Bach2- and Bach1-dependent erythroid lineage 'choice' to cause anemia. Nat Immunol 2018; 19:1059-1070. [PMID: 30250186 DOI: 10.1038/s41590-018-0202-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 07/31/2018] [Indexed: 12/20/2022]
Abstract
Elucidation of how the differentiation of hematopoietic stem and progenitor cells (HSPCs) is reconfigured in response to the environment is critical for understanding the biology and disorder of hematopoiesis. Here we found that the transcription factors (TFs) Bach2 and Bach1 promoted erythropoiesis by regulating heme metabolism in committed erythroid cells to sustain erythroblast maturation and by reinforcing erythroid commitment at the erythro-myeloid bifurcation step. Bach TFs repressed expression of the gene encoding the transcription factor C/EBPβ, as well as that of its target genes encoding molecules important for myelopoiesis and inflammation; they achieved the latter by binding to their regulatory regions also bound by C/EBPβ. Lipopolysaccharide diminished the expression of Bach TFs in progenitor cells and promoted myeloid differentiation. Overexpression of Bach2 in HSPCs promoted erythroid development and inhibited myelopoiesis. Knockdown of BACH1 or BACH2 in human CD34+ HSPCs impaired erythroid differentiation in vitro. Thus, Bach TFs accelerate erythroid commitment by suppressing the myeloid program at steady state. Anemia of inflammation and myelodysplastic syndrome might involve reduced activity of Bach TFs.
Collapse
|
34
|
Guo H, Barberi T, Suresh R, Friedman AD. Progression from the Common Lymphoid Progenitor to B/Myeloid PreproB and ProB Precursors during B Lymphopoiesis Requires C/EBPα. THE JOURNAL OF IMMUNOLOGY 2018; 201:1692-1704. [PMID: 30061199 DOI: 10.4049/jimmunol.1800244] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/11/2018] [Indexed: 11/19/2022]
Abstract
The C/EBPα transcription factor is required for myelopoiesis, with prior observations suggesting additional contributions to B lymphopoiesis. Cebpa expression is evident in common lymphoid progenitor (CLP) and preproB cells but is absent in proB and preB cells. We previously observed that marrow lacking the Cebpa +37 kb enhancer is impaired in producing B cells upon competitive transplantation. Additionally, a Cebpa enhancer/promoter-hCD4 transgene is expressed in B/myeloid CFU. Extending these findings, pan-hematopoietic murine Cebpa enhancer deletion using Mx1-Cre leads to expanded CLP, fewer preproB cells, markedly reduced proB and preB cells, and reduced mature B cells, without affecting T cell numbers. In contrast, enhancer deletion at the proB stage using Mb1-Cre does not impair B cell maturation. Further evaluation of CLP reveals that the Cebpa transgene is expressed almost exclusively in Flt3+ multipotent CLP versus B cell-restricted Flt3- CLP. In vitro, hCD4+ preproB cells produce both B and myeloid cells, whereas hCD4- preproB cells only produce B cells. Additionally, a subset of hCD4- preproB cells express high levels of RAG1-GFP, as seen also in proB cells. Global gene expression analysis indicates that hCD4+ preproB cells express proliferative pathways, whereas B cell development and signal transduction pathways predominate in hCD4- preproB cells. Consistent with these changes, Cebpa enhancer-deleted preproB cells downmodulate cell cycle pathways while upregulating B cell signaling pathways. Collectively, these findings indicate that C/EBPα is required for Flt3+ CLP maturation into preproB cells and then for proliferative Cebpaint B/myeloid preproB cells to progress to Cebpalo B cell-restricted preproB cells and finally to Cebpaneg proB cells.
Collapse
Affiliation(s)
- Hong Guo
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Theresa Barberi
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Rahul Suresh
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Alan D Friedman
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| |
Collapse
|
35
|
Guittard G, Pontarotti P, Granjeaud S, Rodrigues M, Abi-Rached L, Nunès JA. Evolutionary and expression analyses reveal a pattern of ancient duplications and functional specializations in the diversification of the Downstream of Kinase (DOK) genes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 84:193-198. [PMID: 29453999 DOI: 10.1016/j.dci.2018.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/10/2018] [Accepted: 02/10/2018] [Indexed: 06/08/2023]
Abstract
Downstream of Kinase (DOK) proteins represent a multigenic family of adaptors that includes negative regulators of immune cell signaling. Using phylogenetics and intron/exon structure data, we show here that the seven human DOK genes (DOK1 to DOK7) form three highly divergent groups that emerged before the protostome-deuterostome split: DOK1/2/3, DOK4/5/6, and DOK7. For two of these three groups (DOK1/2/3 and DOK4/5/6), further gene duplications occurred in vertebrates and so while chordates only have three DOK genes, vertebrates have seven DOK genes over the three groups. From our expression analysis in humans, we show that each group of DOK genes has a distinct pattern of expression. The DOK1/2/3 group is immune specific, yet each of the three genes in the group has a distinct pattern of expression in immune cells. This immune specificity could thus be ancestral, with the DOK1/2/3 gene also being immune-related in protostomes. The DOK4/5/6 and DOK7 groups represent genes that are much less expressed in immune system than the DOK1/2/3 group. Interestingly, we identify a novel tyrosine based motif that is specific to the vertebrate DOK4/5/6 sequences. The evolution of the DOK genes is thus marked by a pattern of ancient duplications and functional specializations.
Collapse
Affiliation(s)
- Geoffrey Guittard
- Centre de Recherche en Cancérologie de Marseille, Immunity and Cancer Team, Institut Paoli-Calmettes, Inserm, U1068, CNRS, UMR7258, Aix-Marseille Université UM 105, Marseille, France
| | - Pierre Pontarotti
- Aix Marseille Université, CNRS, Centrale Marseille, I2M UMR 7373, équipe évolution Biologique Modélisation, Marseille, France
| | - Samuel Granjeaud
- Centre de Recherche en Cancérologie de Marseille, CiBi Platform, Institut Paoli-Calmettes, Inserm, U1068, CNRS, UMR7258, Aix-Marseille Université UM 105, Marseille, France
| | - Magda Rodrigues
- Centre de Recherche en Cancérologie de Marseille, Immunity and Cancer Team, Institut Paoli-Calmettes, Inserm, U1068, CNRS, UMR7258, Aix-Marseille Université UM 105, Marseille, France
| | - Laurent Abi-Rached
- Equipe ATIP, Aix Marseille Université, CNRS, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France.
| | - Jacques A Nunès
- Centre de Recherche en Cancérologie de Marseille, Immunity and Cancer Team, Institut Paoli-Calmettes, Inserm, U1068, CNRS, UMR7258, Aix-Marseille Université UM 105, Marseille, France.
| |
Collapse
|
36
|
Miura Y, Morooka M, Sax N, Roychoudhuri R, Itoh-Nakadai A, Brydun A, Funayama R, Nakayama K, Satomi S, Matsumoto M, Igarashi K, Muto A. Bach2 Promotes B Cell Receptor-Induced Proliferation of B Lymphocytes and Represses Cyclin-Dependent Kinase Inhibitors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:2882-2893. [PMID: 29540581 DOI: 10.4049/jimmunol.1601863] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 02/20/2018] [Indexed: 12/11/2022]
Abstract
BTB and CNC homology 2 (Bach2) is a transcriptional repressor that is required for the formation of the germinal center (GC) and reactions, including class switch recombination and somatic hypermutation of Ig genes in B cells, within the GC. Although BCR-induced proliferation is essential for GC reactions, the function of Bach2 in regulating B cell proliferation has not been elucidated. In this study, we demonstrate that Bach2 is required to sustain high levels of B cell proliferation in response to BCR signaling. Following BCR engagement in vitro, B cells from Bach2-deficient (Bach2-/-) mice showed lower incorporation of BrdU and reduced cell cycle progression compared with wild-type cells. Bach2-/- B cells also underwent increased apoptosis, as evidenced by an elevated frequency of sub-G1 cells and early apoptotic cells. Transcriptome analysis of BCR-engaged B cells from Bach2-/- mice revealed reduced expression of the antiapoptotic gene Bcl2l1 encoding Bcl-xL and elevated expression of cyclin-dependent kinase inhibitor (CKI) family genes, including Cdkn1a, Cdkn2a, and Cdkn2b Reconstitution of Bcl-xL expression partially rescued the proliferation defect of Bach2-/- B cells. Chromatin immunoprecipitation experiments showed that Bach2 bound to the CKI family genes, indicating that these genes are direct repression targets of Bach2. These findings identify Bach2 as a requisite factor for sustaining high levels of BCR-induced proliferation, survival, and cell cycle progression, and it promotes expression of Bcl-xL and repression of CKI genes. BCR-induced proliferation defects may contribute to the impaired GC formation observed in Bach2-/- mice.
Collapse
Affiliation(s)
- Yuichi Miura
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
- Division of Advanced Surgical Science and Technology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Mizuho Morooka
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Nicolas Sax
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Rahul Roychoudhuri
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, CB22 3AT Cambridge, United Kingdom
| | - Ari Itoh-Nakadai
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Andrey Brydun
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Ryo Funayama
- Department of Cell Proliferation, United Center for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Keiko Nakayama
- Department of Cell Proliferation, United Center for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Susumu Satomi
- Division of Advanced Surgical Science and Technology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; and
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| | - Akihiko Muto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan;
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; and
| |
Collapse
|
37
|
Li J, Shima H, Nishizawa H, Ikeda M, Brydun A, Matsumoto M, Kato H, Saiki Y, Liu L, Watanabe-Matsui M, Iemura K, Tanaka K, Shiraki T, Igarashi K. Phosphorylation of BACH1 switches its function from transcription factor to mitotic chromosome regulator and promotes its interaction with HMMR. Biochem J 2018; 475:981-1002. [PMID: 29459360 DOI: 10.1042/bcj20170520] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 01/31/2018] [Accepted: 02/19/2018] [Indexed: 12/17/2023]
Abstract
The transcription repressor BACH1 performs mutually independent dual roles in transcription regulation and chromosome alignment during mitosis by supporting polar ejection force of mitotic spindle. We now found that the mitotic spindles became oblique relative to the adhesion surface following endogenous BACH1 depletion in HeLa cells. This spindle orientation rearrangement was rescued by re-expression of BACH1 depending on its interactions with HMMR and CRM1, both of which are required for the positioning of mitotic spindle, but independently of its DNA-binding activity. A mass spectrometry analysis of BACH1 complexes in interphase and M phase revealed that BACH1 lost during mitosis interactions with proteins involved in chromatin and gene expression but retained interactions with HMMR and its known partners including CHICA. By analyzing BACH1 modification using stable isotope labeling with amino acids in cell culture, mitosis-specific phosphorylations of BACH1 were observed, and mutations of these residues abolished the activity of BACH1 to restore mitotic spindle orientation in knockdown cells and to interact with HMMR. Detailed histological analysis of Bach1-deficient mice revealed lengthening of the epithelial fold structures of the intestine. These observations suggest that BACH1 performs stabilization of mitotic spindle orientation together with HMMR and CRM1 in mitosis, and that the cell cycle-specific phosphorylation switches the transcriptional and mitotic functions of BACH1.
Collapse
Affiliation(s)
- Jie Li
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Hiroki Shima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University, Sendai 980-8575, Japan
| | - Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Masatoshi Ikeda
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Andrey Brydun
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University, Sendai 980-8575, Japan
| | - Hiroki Kato
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yuriko Saiki
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Liang Liu
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Miki Watanabe-Matsui
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
- Restart Postdoctoral Fellow, Japan Society for the Promotion of Science, Tokyo 102-0083, Japan
| | - Kenji Iemura
- Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Kozo Tanaka
- Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Takuma Shiraki
- Department of Science and Technology on Food Safety, Faculty of Biology-Oriented Science and Technology, Kindai University, 930 Nishimitani, Kinokawashi 649-6493, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
38
|
The mTOR-Bach2 Cascade Controls Cell Cycle and Class Switch Recombination during B Cell Differentiation. Mol Cell Biol 2017; 37:MCB.00418-17. [PMID: 28993481 DOI: 10.1128/mcb.00418-17] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/03/2017] [Indexed: 12/25/2022] Open
Abstract
The transcription factor Bach2 regulates both acquired and innate immunity at multiple steps, including antibody class switching and regulatory T cell development in activated B and T cells, respectively. However, little is known about the molecular mechanisms of Bach2 regulation in response to signaling of cytokines and antigen. We show here that mammalian target of rapamycin (mTOR) controls Bach2 along B cell differentiation with two distinct mechanisms in pre-B cells. First, mTOR complex 1 (mTORC1) inhibited accumulation of Bach2 protein in nuclei and reduced its stability. Second, mTOR complex 2 (mTORC2) inhibited FoxO1 to reduce Bach2 mRNA expression. Using expression profiling and chromatin immunoprecipitation assay, the Ccnd3 gene, encoding cyclin D3, was identified as a new direct target of Bach2. A proper cell cycle was lost at pre-B and mature B cell stages in Bach2-deficient mice. Furthermore, AZD8055, an mTOR inhibitor, increased class switch recombination in wild-type mature B cells but not in Bach2-deficient cells. These results suggest that the mTOR-Bach2 cascade regulates proper cell cycle arrest in B cells as well as immunoglobulin gene rearrangement.
Collapse
|
39
|
Akbari B, Wee P, Yaqubi M, Mohammadnia A. Comprehensive transcriptome mining of the direct conversion of mesodermal cells. Sci Rep 2017; 7:10427. [PMID: 28874788 PMCID: PMC5585404 DOI: 10.1038/s41598-017-10903-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/16/2017] [Indexed: 12/15/2022] Open
Abstract
The direct reprogramming of somatic cells is a promising approach for regenerative medicine, especially in the production of mesoderm layer-derived cells. Meta-analysis studies provide precise insight into the undergoing processes and help increase the efficiency of reprogramming. Here, using 27 high-throughput expression data sets, we analyzed the direct reprogramming of mesodermal cells in humans and mice. Fibroblast-derived cells showed a common expression pattern of up- and down-regulated genes that were mainly involved in the suppression of the fibroblast-specific gene expression program, and may be used as markers of the initiation of reprogramming. Furthermore, we found a specific gene expression profile for each fibroblast-derived cell studied, and each gene set appeared to play specific functional roles in its cell type, suggesting their use as markers for their mature state. Furthermore, using data from protein-DNA interactions, we identified the main transcription factors (TFs) involved in the conversion process and ranked them based on their importance in their gene regulatory networks. In summary, our meta-analysis approach provides new insights on the direct conversion of mesodermal somatic cells, introduces a list of genes as markers for initiation and maturation, and identifies TFs for which manipulating their expression may increase the efficiency of direct conversion.
Collapse
Affiliation(s)
- Bijan Akbari
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ping Wee
- Department of Medical Genetics and Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Moein Yaqubi
- Department of Psychiatry, Sackler Program for Epigenetics and Psychobiology at McGill University, Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada.
| | | |
Collapse
|
40
|
|
41
|
Transcriptional mechanisms that control expression of the macrophage colony-stimulating factor receptor locus. Clin Sci (Lond) 2017; 131:2161-2182. [DOI: 10.1042/cs20170238] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/22/2017] [Accepted: 06/11/2017] [Indexed: 12/17/2022]
Abstract
The proliferation, differentiation, and survival of cells of the macrophage lineage depends upon signals from the macrophage colony-stimulating factor (CSF) receptor (CSF1R). CSF1R is expressed by embryonic macrophages and induced early in adult hematopoiesis, upon commitment of multipotent progenitors to the myeloid lineage. Transcriptional activation of CSF1R requires interaction between members of the E26 transformation-specific family of transcription factors (Ets) (notably PU.1), C/EBP, RUNX, AP-1/ATF, interferon regulatory factor (IRF), STAT, KLF, REL, FUS/TLS (fused in sarcoma/ranslocated in liposarcoma) families, and conserved regulatory elements within the mouse and human CSF1R locus. One element, the Fms-intronic regulatory element (FIRE), within intron 2, is conserved functionally across all the amniotes. Lineage commitment in multipotent progenitors also requires down-regulation of specific transcription factors such as MYB, FLI1, basic leucine zipper transcriptional factor ATF-like (BATF3), GATA-1, and PAX5 that contribute to differentiation of alternative lineages and repress CSF1R transcription. Many of these transcription factors regulate each other, interact at the protein level, and are themselves downstream targets of CSF1R signaling. Control of CSF1R transcription involves feed–forward and feedback signaling in which CSF1R is both a target and a participant; and dysregulation of CSF1R expression and/or function is associated with numerous pathological conditions. In this review, we describe the regulatory network behind CSF1R expression during differentiation and development of cells of the mononuclear phagocyte system.
Collapse
|
42
|
Kurkewich JL, Hansen J, Klopfenstein N, Zhang H, Wood C, Boucher A, Hickman J, Muench DE, Grimes HL, Dahl R. The miR-23a~27a~24-2 microRNA cluster buffers transcription and signaling pathways during hematopoiesis. PLoS Genet 2017; 13:e1006887. [PMID: 28704388 PMCID: PMC5531666 DOI: 10.1371/journal.pgen.1006887] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 07/27/2017] [Accepted: 06/21/2017] [Indexed: 02/07/2023] Open
Abstract
MicroRNA cluster mirn23a has previously been shown to promote myeloid development at the expense of lymphoid development in overexpression and knockout mouse models. This polarization is observed early in hematopoietic development, with an increase in common lymphoid progenitors (CLPs) and a decrease in all myeloid progenitor subsets in adult bone marrow. The pool size of multipotential progenitors (MPPs) is unchanged; however, in this report we observe by flow cytometry that polarized subsets of MPPs are changed in the absence of mirn23a. Additionally, in vitro culture of MPPs and sorted MPP transplants showed that these cells have decreased myeloid and increased lymphoid potential in vitro and in vivo. We investigated the mechanism by which mirn23a regulates hematopoietic differentiation and observed that mirn23a promotes myeloid development of hematopoietic progenitors through regulation of hematopoietic transcription factors and signaling pathways. Early transcription factors that direct the commitment of MPPs to CLPs (Ikzf1, Runx1, Satb1, Bach1 and Bach2) are increased in the absence of mirn23a miRNAs as well as factors that commit the CLP to the B cell lineage (FoxO1, Ebf1, and Pax5). Mirn23a appears to buffer transcription factor levels so that they do not stochastically reach a threshold level to direct differentiation. Intriguingly, mirn23a also inversely regulates the PI3 kinase (PI3K)/Akt and BMP/Smad signaling pathways. Pharmacological inhibitor studies, coupled with dominant active/dominant negative biochemical experiments, show that both signaling pathways are critical to mirn23a’s regulation of hematopoietic differentiation. Lastly, consistent with mirn23a being a physiological inhibitor of B cell development, we observed that the essential B cell transcription factor EBF1 represses expression of mirn23a. In summary, our data demonstrates that mirn23a regulates a complex array of transcription and signaling pathways to modulate adult hematopoiesis. MicroRNAs (miRNAs) are small ~22 nucleotide long RNA molecules that are involved in regulating multiple cellular processes through inhibiting the expression of target proteins. We previously identified a gene (mirn23a) that codes for 3 miRNAs that control the development of immune cells in the bone marrow. The miRNAs promote the development of innate immune cells, macrophages and granulocytes, while repressing the development of B cells. Here we show that mirn23a miRNAs negatively affect the expression of multiple proteins that are involved in directing blood progenitor cells to become B cells. Additionally, we observed that modulation of FoxO1 and Smad proteins, downstream effectors of two signaling pathways (PI3 kinase/ Akt and BMP/ Smad), is critical to direct immune cell development. This is the first observation that these pathways are potentially coregulated during the commitment of blood progenitors to mature cells of the immune system. Consistent with mirn23a being a critical gene for committing progenitors to innate immune cells at the expense of B cells, we observed that a critical B cell protein represses the expression of mirn23a. In conclusion, we demonstrate the mirn23a regulation of blood development is due to a complex regulation of both transcription factors and signaling pathways.
Collapse
Affiliation(s)
- Jeffrey L. Kurkewich
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
- Harper Cancer Research Institute, South Bend, IN, United States of America
| | - Justin Hansen
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
- Harper Cancer Research Institute, South Bend, IN, United States of America
| | - Nathan Klopfenstein
- Harper Cancer Research Institute, South Bend, IN, United States of America
- Department of Microbiology and Immunology, Indiana University School of Medicine, South Bend, IN, United States of America
| | - Helen Zhang
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
- Harper Cancer Research Institute, South Bend, IN, United States of America
| | - Christian Wood
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
- Harper Cancer Research Institute, South Bend, IN, United States of America
| | - Austin Boucher
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
- Harper Cancer Research Institute, South Bend, IN, United States of America
| | - Joseph Hickman
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
- Harper Cancer Research Institute, South Bend, IN, United States of America
| | - David E. Muench
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - H. Leighton Grimes
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Richard Dahl
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
- Harper Cancer Research Institute, South Bend, IN, United States of America
- Department of Microbiology and Immunology, Indiana University School of Medicine, South Bend, IN, United States of America
- * E-mail:
| |
Collapse
|
43
|
Igarashi K, Kurosaki T, Roychoudhuri R. BACH transcription factors in innate and adaptive immunity. Nat Rev Immunol 2017; 17:437-450. [PMID: 28461702 DOI: 10.1038/nri.2017.26] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BTB and CNC homology (BACH) proteins are transcriptional repressors of the basic region leucine zipper (bZIP) transcription factor family. Recent studies indicate widespread roles of BACH proteins in controlling the development and function of the innate and adaptive immune systems, including the differentiation of effector and memory cells of the B and T cell lineages, CD4+ regulatory T cells and macrophages. Here, we emphasize similarities at a molecular level in the cell-type-specific activities of BACH factors, proposing that competitive interactions of BACH proteins with transcriptional activators of the bZIP family form a common mechanistic theme underlying their diverse actions. The findings contribute to a general understanding of how transcriptional repressors shape lineage commitment and cell-type-specific functions through repression of alternative lineage programmes.
Collapse
Affiliation(s)
- Kazuhiko Igarashi
- Department of Biochemistry, Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Tomohiro Kurosaki
- Laboratory for Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi-ku, Yokohama 230-0045, Japan
| | - Rahul Roychoudhuri
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Cambridge CB22 3AT, UK
| |
Collapse
|
44
|
Sato Y, Kato H, Ebina-Shibuya R, Itoh-Nakadai A, Okuyama R, Igarashi K. Bach2 Controls Homeostasis of Eosinophils by Restricting the Type-2 Helper Function of T Cells. TOHOKU J EXP MED 2017; 241:175-182. [PMID: 28216546 DOI: 10.1620/tjem.241.175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Bach2 is a transcription factor which represses its target genes and plays important roles in the differentiation of B and T lymphoid cells. Bach2-deficient (KO) mice develop severe pulmonary alveolar proteinosis, which is associated with increased numbers of granulocytes and T cells. Bach2 is essential for the regulation of T cells, but its role in the regulation of granulocytes is not clear. Here, we observed increased numbers of eosinophils but not neutrophils in the bone marrow, spleen, peripheral blood, and bronchoalveolar lavage fluids of Bach2 KO mice compared with those of wild-type (WT) mice. Upon co-transplantation of the bone marrow cells from CD45.2 Bach2 KO and CD45.1/CD45.2 double-positive WT mice to irradiated WT CD45.1/CD45.2 mice, the reconstituted numbers of eosinophils were similar between Bach2 KO and WT cells. These results showed that the deficiency of Bach2 in eosinophils did not directly drive the differentiation of eosinophils. To investigate the effect of Bach2 KO CD4+ T cells upon eosinophils, we analyzed Rag2/Bach2-double deficient (dKO) mice which lack lymphocytes including CD4+ T cells. Rag2/Bach2 dKO mice did not show any increase in the numbers of eosinophils. Importantly, Bach2 KO mice showed an increase of interleukin-5 (Il-5) in the sera compared with WT mice. These results suggest that up-regulated functions of CD4+ T cells including secretion of Il-5 resulted in proliferation and/or migration to peripheral tissues of eosinophils in Bach2 KO mice. We propose that Bach2 controls homeostasis of eosinophils via restricting the production of Il-5 in CD4+ T cells.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Biochemistry, Tohoku University Graduate School of Medicine
| | | | | | | | | | | |
Collapse
|