1
|
Chen W, Chen X, Yao L, Feng J, Li F, Shan Y, Ren L, Zhuo C, Feng M, Zhong S, He C. A global view of altered ligand-receptor interactions in bone marrow aging based on single-cell sequencing. Comput Struct Biotechnol J 2024; 23:2754-2762. [PMID: 39050783 PMCID: PMC11267010 DOI: 10.1016/j.csbj.2024.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
Altered cell-cell communication is a hallmark of aging, but its impact on bone marrow aging remains poorly understood. Based on a common and effective pipeline and single-cell transcriptome sequencing, we detected 384,124 interactions including 2575 ligand-receptor pairs and 16 non-adherent bone marrow cell types in old and young mouse and identified a total of 5560 significantly different interactions, which were then verified by flow cytometry and quantitative real-time PCR. These differential ligand-receptor interactions exhibited enrichment for the senescence-associated secretory phenotypes. Further validation demonstrated supplementing specific extracellular ligands could modify the senescent signs of hematopoietic stem cells derived from old mouse. Our work provides an effective procedure to detect the ligand-receptor interactions based on single-cell sequencing, which contributes to understand mechanisms and provides a potential strategy for intervention of bone marrow aging.
Collapse
Affiliation(s)
- Wenbo Chen
- School of Basic Medical Sciences, Taikang Medical School, Wuhan University, Wuhan 430071, China
| | - Xin Chen
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Lei Yao
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Jing Feng
- School of Computer Science, Wuhan University, Wuhan 430072, China
| | - Fengyue Li
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Yuxin Shan
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Linli Ren
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Chenjian Zhuo
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Mingqian Feng
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Shan Zhong
- School of Basic Medical Sciences, Taikang Medical School, Wuhan University, Wuhan 430071, China
| | - Chunjiang He
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
2
|
Madel MB, Ibáñez L, Ciucci T, Halper J, Boutin A, Beldi G, Lavanant AC, Garchon HJ, Rouleau M, Mueller CG, Peyrin-Biroulet L, Moulin D, Blin-Wakkach C, Wakkach A. Dysregulated myeloid differentiation in colitis is induced by inflammatory osteoclasts in a TNFα-dependent manner. Mucosal Immunol 2024:S1933-0219(24)00098-9. [PMID: 39332768 DOI: 10.1016/j.mucimm.2024.09.005] [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: 10/27/2023] [Revised: 09/14/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Inflammatory bowel disease (IBD) is characterized by very severe intestinal inflammation associated with extra-intestinal manifestations. One of the most critical ones is bone destruction, which remains a major cause of morbidity and a risk factor for osteopenia and osteoporosis in IBD patients. In various mouse models of IBD, we and other have demonstrated concomitant bone loss due to a significant increase in osteoclast activity. Besides bone resorption, osteoclasts are known to control hematopoietic niches in vivo and modulate inflammatory responses in vitro, suggesting they may participate in chronic inflammation in vivo. Here, using different models of colitis, we showed that osteoclast inhibition significantly reduced disease severity and that induction of osteoclast differentiation by RANKL contributed to disease worsening. Our results demonstrate a direct link between osteoclast activity and myeloid cell accumulation in the intestine during colitis. RNAseq analysis of osteoclasts from colitic mice revealed overexpression of genes involved in the remodeling of hematopoietic stem cell niches. We also demonstrated that osteoclasts induced hematopoietic progenitor proliferation accompanied by a myeloid skewing in the early phases of colitis, which was confirmed in a model of RANKL-induced osteoclastogenesis. Mechanistically, inhibition of TNF-α reduced the induction of myeloid skewing by OCL both in vitro and in vivo. Lastly, we observed that osteoclastic activity and the proportion of myeloid cells in the blood are positively correlated in patients with Crohn's disease. Collectively, our results shed light on a new role of osteoclasts in colitis in vivo, demonstrating they exert their colitogenic activity through an early action on hematopoiesis, leading to an increase in myelopoiesis sustaining gut inflammation.
Collapse
Affiliation(s)
| | - Lidia Ibáñez
- Université Côte d'Azur, CNRS, LP2M, Nice, France
| | - Thomas Ciucci
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Julia Halper
- Université Côte d'Azur, CNRS, LP2M, Nice, France
| | | | - Ghada Beldi
- Université Côte d'Azur, CNRS, LP2M, Nice, France
| | - Alice C Lavanant
- CNRS UPR 3572, IBMC, University of Strasbourg, 67000 Strasbourg, France
| | - Henri-Jean Garchon
- Université Paris-Saclay, UVSQ, INSERM, Infection et inflammation, 78180 Montigny-Le-Bretonneux, France
| | | | | | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, IHU INFINY, CHRU Nancy, F-54500 Vandœuvre-lès-Nancy, France; Université de Lorraine, INSERM, NGERE, F-54500 Vandoeuvre les Nancy, France
| | - David Moulin
- Université de Lorraine, CNRS, IMoPA, F-54500 Vandœuvre Les Nancy, France; IHU INFINY, Contrat d'interface, CHRU Nancy, France
| | | | | |
Collapse
|
3
|
Zhi Y, Qiu W, Tian G, Song S, Zhao W, Du X, Sun X, Chen Y, Huang H, Li J, Yu Y, Li M, Lv G. Donor and recipient hematopoietic stem and progenitor cells mobilization in liver transplantation patients. Stem Cell Res Ther 2024; 15:231. [PMID: 39075608 PMCID: PMC11288126 DOI: 10.1186/s13287-024-03855-5] [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: 05/24/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Hematopoietic stem and progenitor cells (HSPCs) mobilize from bone marrow to peripheral blood in response to stress. The impact of alloresponse-induced stress on HSPCs mobilization in human liver transplantation (LTx) recipients remains under-investigated. METHODS Peripheral blood mononuclear cell (PBMC) samples were longitudinally collected from pre- to post-LTx for one year from 36 recipients with acute rejection (AR), 74 recipients without rejection (NR), and 5 recipients with graft-versus-host disease (GVHD). 28 PBMC samples from age-matched healthy donors were collected as healthy control (HC). Multi-color flow cytometry (MCFC) was used to immunophenotype HSPCs and their subpopulations. Donor recipient-distinguishable major histocompatibility complex (MHC) antibodies determined cell origin. RESULTS Before LTx, patients who developed AR after transplant contained more HSPCs in PBMC samples than HC, while the NR group patients contained fewer HSPCs than HC. After LTx, the HSPC ratio in the AR group sharply decreased and became less than HC within six months, and dropped to a comparable NR level afterward. During the one-year follow-up period, myeloid progenitors (MPs) biased differentiation was observed in all LTx recipients who were under tacrolimus-based immunosuppressive treatment. During both AR and GVHD episodes, the recipient-derived and donor-derived HSPCs mobilized into the recipient's blood-circulation and migrated to the target tissue, respectively. The HSPCs percentage in blood reduced after the disease was cured. CONCLUSIONS A preoperative high HSPC ratio in blood characterizes recipients who developed AR after LTx. Recipients exhibited a decline in blood-circulating HSPCs after transplant, the cells mobilized into the blood and migrated to target tissue during alloresponse.
Collapse
Affiliation(s)
- Yao Zhi
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Wei Qiu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Guangyao Tian
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Shifei Song
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Wenchao Zhao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xiaodong Du
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xiaodong Sun
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yuguo Chen
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Heyu Huang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jing Li
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Ying Yu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Mingqian Li
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
4
|
Alves R, Pires A, Jorge J, Balça-Silva J, Gonçalves AC, Sarmento-Ribeiro AB. Batimastat Induces Cytotoxic and Cytostatic Effects in In Vitro Models of Hematological Tumors. Int J Mol Sci 2024; 25:4554. [PMID: 38674139 PMCID: PMC11050270 DOI: 10.3390/ijms25084554] [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: 03/31/2024] [Revised: 04/16/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024] Open
Abstract
The role of metalloproteinases (MMPs) in hematological malignancies, like acute myeloid leukemia (AML), myelodysplastic neoplasms (MDS), and multiple myeloma (MM), is well-documented, and these pathologies remain with poor outcomes despite treatment advancements. In this study, we investigated the effects of batimastat (BB-94), an MMP inhibitor (MMPi), in single-administration and daily administration schemes in AML, MDS, and MM cell lines. We used four hematologic neoplasia cell lines: the HL-60 and NB-4 cells as AML models, the F36-P cells as an MDS model, and the H929 cells as a model of MM. We also tested batimastat toxicity in a normal human lymphocyte cell line (IMC cells). BB-94 decreases cell viability and density in a dose-, time-, administration-scheme-, and cell-line-dependent manner, with the AML cells displaying higher responses. The efficacy in inducing apoptosis and cell cycle arrests is dependent on the cell line (higher effects in AML cells), especially with lower daily doses, which may mitigate treatment toxicity. Furthermore, BB-94 activated apoptosis via caspases and ERK1/2 pathways. These findings highlight batimastat's therapeutic potential in hematological malignancies, with daily dosing emerging as a strategy to minimize adverse effects.
Collapse
Affiliation(s)
- Raquel Alves
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.P.); (J.J.); (J.B.-S.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
| | - Ana Pires
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.P.); (J.J.); (J.B.-S.); (A.B.S.-R.)
- HistologiX, BioCity, Innovation, Pennyfoot St., Nottingham NG1 1GF, UK
| | - Joana Jorge
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.P.); (J.J.); (J.B.-S.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
| | - Joana Balça-Silva
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.P.); (J.J.); (J.B.-S.); (A.B.S.-R.)
- NOVA Medical School, New University of Lisbon, 1150-090 Lisbon, Portugal
| | - Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.P.); (J.J.); (J.B.-S.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.P.); (J.J.); (J.B.-S.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Hematology Service, Centro Hospitalar Universitário de Coimbra, Unidade Local de Saúde de Coimbra, 3000-061 Coimbra, Portugal
| |
Collapse
|
5
|
Wu J, Ma L, Gong Q, Chen Y, Chen L, Shi C. NEAR-INFRARED DYE IR-780 ALLEVIATES HEMATOPOIETIC SYSTEM DAMAGE BY PROMOTING HEMATOPOIETIC STEM CELLS INTO QUIESCENCE. Shock 2024; 61:442-453. [PMID: 38411611 DOI: 10.1097/shk.0000000000002317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
ABSTRACT Potential radiation exposure is a general concern, but there still lacks radioprotective countermeasures. Here, we found a small molecular near-infrared dye IR-780, which promoted hematopoietic stem cells (HSCs) into quiescence to resist stress. When mice were treated with IR-780 before stress, increased HSC quiescence and better hematopoietic recovery were observed in mice in stress conditions. However, when given after radiation, IR-780 did not show obvious benefit. Transplantation assay and colony-forming assay were carried out to determine self-renewal ability and repopulation capacity of HSCs. Furthermore, IR-780 pretreatment reduced the generation of reactive oxygen species (ROS) and DNA damage in HSCs after radiation. In homeostasis, the percentage of Lineage - , Sca-1 + , and c-Kit + cells and long-term HSCs (LT-HSCs) were improved, and more HSCs were in G0 state after administration of IR-780. Further investigations showed that IR-780 selectively accumulated in mitochondria membrane potential high LT-HSCs (MMP-high LT-HSCs). Finally, IR-780 promoted human CD34 + HSC reconstruction ability in NOD-Prkdc scid Il2rg null mice after transplantation and improved repopulation capacity in vitro culture. Our research showed that IR-780 selectively entered MMP-high LT-HSCs and promoted them into dormancy, thus reducing hematopoietic injury and improving regeneration capacity. This novel approach might hold promise as a potential countermeasure for radiation injury.
Collapse
Affiliation(s)
- Jie Wu
- Institute of Rocket Force Medicine, State Key of Trauma and Chemical Poisoning Third Military Medical University (Army Medical University), Chongqing, China
| | - Le Ma
- Institute of Rocket Force Medicine, State Key of Trauma and Chemical Poisoning Third Military Medical University (Army Medical University), Chongqing, China
| | - Qiang Gong
- Department of Hematology, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Yan Chen
- Institute of Rocket Force Medicine, State Key of Trauma and Chemical Poisoning Third Military Medical University (Army Medical University), Chongqing, China
| | - Long Chen
- Institute of Rocket Force Medicine, State Key of Trauma and Chemical Poisoning Third Military Medical University (Army Medical University), Chongqing, China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key of Trauma and Chemical Poisoning Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
6
|
Berke Mentese I, Kronbichler A. Inhibition of clonal expansion of parietal epithelial cells and crescent-podocyte transition in severe glomerulonephritis: on the way to targeted therapy? Clin Kidney J 2023; 16:1057-1058. [PMID: 37398681 PMCID: PMC10310498 DOI: 10.1093/ckj/sfad054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Indexed: 07/04/2023] Open
|
7
|
Hao X, Shen Y, Chen N, Zhang W, Valverde E, Wu L, Chan HL, Xu Z, Yu L, Gao Y, Bado I, Michie LN, Rivas CH, Dominguez LB, Aguirre S, Pingel BC, Wu YH, Liu F, Ding Y, Edwards DG, Liu J, Alexander A, Ueno NT, Hsueh PR, Tu CY, Liu LC, Chen SH, Hung MC, Lim B, Zhang XHF. Osteoprogenitor-GMP crosstalk underpins solid tumor-induced systemic immunosuppression and persists after tumor removal. Cell Stem Cell 2023; 30:648-664.e8. [PMID: 37146584 PMCID: PMC10165729 DOI: 10.1016/j.stem.2023.04.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/09/2023] [Accepted: 04/06/2023] [Indexed: 05/07/2023]
Abstract
Remote tumors disrupt the bone marrow (BM) ecosystem (BME), eliciting the overproduction of BM-derived immunosuppressive cells. However, the underlying mechanisms remain poorly understood. Herein, we characterized breast and lung cancer-induced BME shifts pre- and post-tumor removal. Remote tumors progressively lead to osteoprogenitor (OP) expansion, hematopoietic stem cell dislocation, and CD41- granulocyte-monocyte progenitor (GMP) aggregation. The tumor-entrained BME is characterized by co-localization between CD41- GMPs and OPs. OP ablation abolishes this effect and diminishes abnormal myeloid overproduction. Mechanistically, HTRA1 carried by tumor-derived small extracellular vesicles upregulates MMP-13 in OPs, which in turn induces the alterations in the hematopoietic program. Importantly, these effects persist post-surgery and continue to impair anti-tumor immunity. Conditional knockout or inhibition of MMP-13 accelerates immune reinstatement and restores the efficacies of immunotherapies. Therefore, tumor-induced systemic effects are initiated by OP-GMP crosstalk that outlasts tumor burden, and additional treatment is required to reverse these effects for optimal therapeutic efficacy.
Collapse
Affiliation(s)
- Xiaoxin Hao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yichao Shen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Nan Chen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Weijie Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Elizabeth Valverde
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Ling Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hilda L Chan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhan Xu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Liqun Yu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yang Gao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Igor Bado
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Laura Natalee Michie
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Charlotte Helena Rivas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Luis Becerra Dominguez
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Immunology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Sergio Aguirre
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Bradley C Pingel
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Immunology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yi-Hsuan Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yunfeng Ding
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - David G Edwards
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jun Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Angela Alexander
- Department of Breast Medical Oncology and Morgan Welch IBC Research Program and Clinic, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology and Morgan Welch IBC Research Program and Clinic, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; University of Hawai'i Cancer Center (UHCC), 701 Ilalo Street, Honolulu, HI 96813, USA
| | - Po-Ren Hsueh
- Departments of Laboratory Medicine and Internal Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Chih-Yen Tu
- School of Medicine, College of Medicine, China Medical University, Taichung 406, Taiwan; Division of Pulmonary and Critical Care, Department of Internal Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Liang-Chih Liu
- School of Medicine, College of Medicine, China Medical University, Taichung 406, Taiwan; Division of Breast Surgery, Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Shu-Hsia Chen
- Immunomonitoring Core, Center for Immunotherapy Research, Houston Methodist Research Institute (HMRI), Houston, TX, USA
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung 40402, Taiwan
| | - Bora Lim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
8
|
Lim VY, Feng X, Miao R, Zehentmeier S, Ewing-Crystal N, Lee M, Tumanov AV, Oh JE, Iwasaki A, Wang A, Choi J, Pereira JP. Mature B cells and mesenchymal stem cells control emergency myelopoiesis. Life Sci Alliance 2023; 6:e202301924. [PMID: 36717247 PMCID: PMC9889502 DOI: 10.26508/lsa.202301924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
Systemic inflammation halts lymphopoiesis and prioritizes myeloid cell production. How blood cell production switches from homeostasis to emergency myelopoiesis is incompletely understood. Here, we show that lymphotoxin-β receptor (LTβR) signaling in combination with TNF and IL-1 receptor signaling in bone marrow mesenchymal stem cells (MSCs) down-regulates Il7 expression to shut down lymphopoiesis during systemic inflammation. LTβR signaling in MSCs also promoted CCL2 production during systemic inflammation. Pharmacological or genetic blocking of LTβR signaling in MSCs partially enabled lymphopoiesis and reduced monocyte numbers in the spleen during systemic inflammation, which correlated with reduced survival during systemic bacterial and viral infections. Interestingly, lymphotoxin-α1β2 delivered by B-lineage cells, and specifically by mature B cells, contributed to promote Il7 down-regulation and reduce MSC lymphopoietic activity. Our studies revealed an unexpected role of LTβR signaling in MSCs and identified recirculating mature B cells as an important regulator of emergency myelopoiesis.
Collapse
Affiliation(s)
- Vivian Y Lim
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Xing Feng
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Runfeng Miao
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Sandra Zehentmeier
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Nathan Ewing-Crystal
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Moonyoung Lee
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
| | - Alexei V Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ji Eun Oh
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Andrew Wang
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Medicine (Rheumatology), School of Medicine, Yale University, New Haven, CT, USA
| | - Jungmin Choi
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA
| | - João P Pereira
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
9
|
Liu Y, Liu Z, Hu L, He L, Yang L, Qin Z, Xie Y, Peng X, Dai L. Function of stem cells in radiation-induced damage. Int J Radiat Biol 2023; 99:1483-1494. [PMID: 36912588 DOI: 10.1080/09553002.2023.2188935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/27/2023] [Indexed: 03/14/2023]
Abstract
PURPOSE The aim of this review is to discuss previous studies on the function of stem cells in radiation-induced damage, and the factors affecting these processes, in the hope of improving our understanding of the different stem cells and the communication networks surrounding them. This is essential for the development of effective stem cell-based therapies to regenerate or replace normal tissues damaged by radiation. CONCLUSION In salivary glands, senescence-associated cytokines and inflammation-associated cells have a greater effect on stem cells. In the intestinal glands, Paneth cells strongly affect stem cell-mediated tissue regeneration after radiation treatment. In the pancreas, β-cells as well as protein C receptor positive (Procr) cells are expected to be key cells in the treatment of diabetes. In the bone marrow, a variety of cytokines such as CXC-chemokine ligand 12 (CXCL12) and stem cell factor (SCF), contribute to the functional recovery of hematopoietic stem cells after irradiation.
Collapse
Affiliation(s)
- Yingtong Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zheran Liu
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Liqiang Hu
- West China-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Ling He
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lianlian Yang
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zijian Qin
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yuping Xie
- Department of Oncology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lei Dai
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Zhang H, Liesveld JL, Calvi LM, Lipe BC, Xing L, Becker MW, Schwarz EM, Yeh SCA. The roles of bone remodeling in normal hematopoiesis and age-related hematological malignancies. Bone Res 2023; 11:15. [PMID: 36918531 PMCID: PMC10014945 DOI: 10.1038/s41413-023-00249-w] [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: 08/01/2022] [Revised: 12/24/2022] [Accepted: 01/26/2023] [Indexed: 03/16/2023] Open
Abstract
Prior research establishing that bone interacts in coordination with the bone marrow microenvironment (BMME) to regulate hematopoietic homeostasis was largely based on analyses of individual bone-associated cell populations. Recent advances in intravital imaging has suggested that the expansion of hematopoietic stem cells (HSCs) and acute myeloid leukemia cells is restricted to bone marrow microdomains during a distinct stage of bone remodeling. These findings indicate that dynamic bone remodeling likely imposes additional heterogeneity within the BMME to yield differential clonal responses. A holistic understanding of the role of bone remodeling in regulating the stem cell niche and how these interactions are altered in age-related hematological malignancies will be critical to the development of novel interventions. To advance this understanding, herein, we provide a synopsis of the cellular and molecular constituents that participate in bone turnover and their known connections to the hematopoietic compartment. Specifically, we elaborate on the coupling between bone remodeling and the BMME in homeostasis and age-related hematological malignancies and after treatment with bone-targeting approaches. We then discuss unresolved questions and ambiguities that remain in the field.
Collapse
Affiliation(s)
- Hengwei Zhang
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Jane L Liesveld
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Laura M Calvi
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Endocrinology/Metabolism, University of Rochester Medical Center, Rochester, NY, USA
| | - Brea C Lipe
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Michael W Becker
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Allergy/Immunology/Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Shu-Chi A Yeh
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.
- Department of Physiology/Pharmacology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
11
|
Wang G, Romero Y, Thevarajan I, Zolkiewska A. ADAM12 abrogation alters immune cell infiltration and improves response to checkpoint blockade therapy in the T11 murine model of triple-negative breast cancer. Oncoimmunology 2022; 12:2158006. [PMID: 36545255 PMCID: PMC9762783 DOI: 10.1080/2162402x.2022.2158006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Immunosuppressive tumor microenvironment (TME) impedes anti-tumor immune responses and contributes to immunotherapy resistance in triple-negative breast cancer (TNBC). ADAM12, a member of cell surface metalloproteases, is selectively upregulated in mesenchymal/claudin-low TNBCs, where its expression is largely restricted to tumor cells. The role of cancer cell-expressed ADAM12 in modulating the immune TME is not known. We show that Adam12 knockout in the T11 mouse syngeneic transplantation model of claudin-low TNBC leads to decreased numbers of tumor-infiltrating neutrophils (TINs)/polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) and increased numbers of tumor-infiltrating B cells and T cells. ADAM12 loss in cancer cells increases chemotaxis of B cells in vitro and this effect is eliminated by inhibition of CXCR4, a receptor for CXCL12, or anti-CXCL12 blocking antibody. Importantly, ADAM12 loss in T11 cancer cells sensitizes tumors to anti-PD1/anti-CTLA4 combination therapy, although the initial responsiveness is followed by acquired therapy resistance. Depletion of B cells in mice eliminates the improved response to immune checkpoint blockade of Adam12 knockout T11 tumors. Analysis of gene expression data for claudin-low TNBCs from the METABRIC patient cohort shows significant inverse correlations between ADAM12 and gene expression signatures of several anti-tumor immune cell populations, as well as a significant positive correlation between ADAM12 and gene expression signature of TINs/PMN-MDSCs. Collectively, these results implicate ADAM12 in immunosuppression within the TME in TNBC.
Collapse
Affiliation(s)
- Guanpeng Wang
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, USA
| | - Yeni Romero
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, USA
| | - Indhujah Thevarajan
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, USA
| | - Anna Zolkiewska
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, USA,CONTACT Anna Zolkiewska Department of Biochemistry and molecular Biophysics, Kansas State University, 141 Chalmers Hall, Manhattan, KS66506, USA
| |
Collapse
|
12
|
Wang Q, Wang K, Tan X, Li Z, Wang H. Immunomodulatory role of metalloproteases in cancers: Current progress and future trends. Front Immunol 2022; 13:1064033. [PMID: 36591235 PMCID: PMC9800621 DOI: 10.3389/fimmu.2022.1064033] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Metalloproteinases (MPs) is a large family of proteinases with metal ions in their active centers. According to the different domains metalloproteinases can be divided into a variety of subtypes mainly including Matrix Metalloproteinases (MMPs), A Disintegrin and Metalloproteases (ADAMs) and ADAMs with Thrombospondin Motifs (ADAMTS). They have various functions such as protein hydrolysis, cell adhesion and remodeling of extracellular matrix. Metalloproteinases expressed in multiple types of cancers and participate in many pathological processes involving tumor genesis and development, invasion and metastasis by regulating signal transduction and tumor microenvironment. In this review, based on the current research progress, we summarized the structure of MPs, their expression and especially immunomodulatory role and mechanisms in cancers. Additionally, a relevant and timely update of recent advances and future directions were provided for the diagnosis and immunotherapy targeting MPs in cancers.
Collapse
Affiliation(s)
- Qi Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Kai Wang
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Xiaojing Tan
- Department of Oncology, Dongying People's Hospital, Dongying, China
| | - Zhenxiang Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China,*Correspondence: Zhenxiang Li, ; Haiyong Wang,
| | - Haiyong Wang
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China,*Correspondence: Zhenxiang Li, ; Haiyong Wang,
| |
Collapse
|
13
|
Azar A, Bhutta MF, Del-Pozo J, Milne E, Cheeseman M. Trans-cortical vessels in the mouse temporal bulla bone are a means to recruit myeloid cells in chronic otitis media and limit peripheral leukogram changes. Front Genet 2022; 13:985214. [PMID: 36246635 PMCID: PMC9555619 DOI: 10.3389/fgene.2022.985214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic otitis media, inflammation of the middle ear, is a sequel to acute otitis media in ∼8% of children. Chronic otitis media with effusion is the most common cause of childhood deafness and is characterised by effusion of white blood cells into the auditory bulla cavity. Skull flat bones have trans-cortical vessels which are responsible for the majority of blood flow in and out of the bone. In experimental models of stroke and aseptic meningitis there is preferential recruitment of myeloid cells (neutrophils and monocytes) from the marrow in skull flat bones. We report trans-cortical vessels in the mouse temporal bone connect to the bulla mucosal vasculature and potentially represent a means to recruit myeloid cells directly into the inflamed bulla. The mutant mouse strains Junbo (MecomJbo/+) and Jeff (Fbxo11Jf/+) develop chronic otitis spontaneously; MecomJbo/+ mice have highly cellular neutrophil (90%) rich bulla exudates whereas Fbxo11Jf/+ mice have low cellularity serous effusions (5% neutrophils) indicating differing demand for neutrophil recruitment. However we found peripheral leukograms of MecomJbo/+ and Fbxo11Jf/+ mice are similar to their respective wild-type littermate controls with healthy bullae and infer preferential mobilization of myeloid cells from temporal bulla bone marrow may mitigate the need for a systemic inflammatory reaction. The cytokines, chemokines and haematopoietic factors found in the inflamed bulla represent candidate signalling molecules for myeloid cell mobilization from temporal bone marrow. The density of white blood cells in the bulla cavity is positively correlated with extent of mucosal thickening in MecomJbo/+, Fbxo11Jf/+, and EdaTa mice and is accompanied by changes in epithelial populations and bone remodelling. In MecomJbo/+ mice there was a positive correlation between bulla cavity WBC numbers and total bacterial load. The degree of inflammation varies between contralateral bullae and between mutant mice of different ages suggesting inflammation may wax and wane and may be re-initiated by a new wave of bacterial infection. Clearance of white blood cells and inflammatory stimuli from the bulla cavity is impaired and this may create a pro-inflammatory feedback loop which further exacerbates otitis media and delays its resolution.
Collapse
Affiliation(s)
- Ali Azar
- Developmental Biology Division, Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Mahmood F. Bhutta
- Brighton and Sussex Medical School, Brighton, United Kingdom
- Department of ENT, Royal Sussex County Hospital, Brighton, United Kingdom
| | - Jorge Del-Pozo
- Veterinary Pathology, The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Elspeth Milne
- Veterinary Pathology, The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Michael Cheeseman
- Developmental Biology Division, Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Comparative Pathology, Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- *Correspondence: Michael Cheeseman,
| |
Collapse
|
14
|
Lim R, Banerjee A, Biswas R, Chari AN, Raghavan S. Mechanotransduction through adhesion molecules: Emerging roles in regulating the stem cell niche. Front Cell Dev Biol 2022; 10:966662. [PMID: 36172276 PMCID: PMC9511051 DOI: 10.3389/fcell.2022.966662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
Stem cells have been shown to play an important role in regenerative medicine due to their proliferative and differentiation potential. The challenge, however, lies in regulating and controlling their potential for this purpose. Stem cells are regulated by growth factors as well as an array of biochemical and mechanical signals. While the role of biochemical signals and growth factors in regulating stem cell homeostasis is well explored, the role of mechanical signals has only just started to be investigated. Stem cells interact with their niche or to other stem cells via adhesion molecules that eventually transduce mechanical cues to maintain their homeostatic function. Here, we present a comprehensive review on our current understanding of the influence of the forces perceived by cell adhesion molecules on the regulation of stem cells. Additionally, we provide insights on how this deeper understanding of mechanobiology of stem cells has translated toward therapeutics.
Collapse
Affiliation(s)
- Ryan Lim
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Avinanda Banerjee
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Ritusree Biswas
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
- Sastra University, Thanjavur, TN, India
| | - Anana Nandakumar Chari
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Srikala Raghavan
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
| |
Collapse
|
15
|
Pastrana-Otero I, Majumdar S, Gilchrist AE, Harley BAC, Kraft ML. Identification of the Differentiation Stages of Living Cells from the Six Most Immature Murine Hematopoietic Cell Populations by Multivariate Analysis of Single-Cell Raman Spectra. Anal Chem 2022; 94:11999-12007. [PMID: 36001072 PMCID: PMC9628127 DOI: 10.1021/acs.analchem.2c00714] [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] [Indexed: 11/29/2022]
Abstract
Efforts to expand hematopoietic stem and progenitor cells (HSPCs) in vitro are motivated by their use in the treatment of leukemias and other blood and immune system diseases. The combinations of extrinsic cues within the hematopoietic stem cell (HSC) niche that lead to HSC fate decisions remain unknown. New noninvasive and location-specific techniques are needed to enable identification of the differentiation stages of individual hematopoietic cells on biomaterial microarray screening platforms that minimize the usage of rare HSCs. Here, we show that a combination of Raman microspectroscopy and partial least-squares discriminant analysis (PLS-DA) enables the location-specific identification of individual living cells from the six most immature hematopoietic cell populations, HSC, multipotent progenitor (MPP)-1, MPP-2, MPP-3, common myeloid progenitor, and common lymphoid progenitor. Better than 90% accuracy was achieved. We show that the accuracy of this differentiation stage identification was based on spectral features associated with cell biochemistries. This work establishes that PLS-DA can capture the subtle spectral variations between as many as six closely related cell populations in the presence of potentially significant within-population spectral variation. This noninvasive approach can be used to screen HSC fate decisions elicited by extrinsic cues within biomaterial microarray screening platforms.
Collapse
Affiliation(s)
- Isamar Pastrana-Otero
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Sayani Majumdar
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Aidan E Gilchrist
- Department of Materials Science and Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Brendan A C Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Mary L Kraft
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
16
|
Melica ME, Antonelli G, Semeraro R, Angelotti ML, Lugli G, Landini S, Ravaglia F, La Regina G, Conte C, De Chiara L, Peired AJ, Mazzinghi B, Donati M, Molli A, Steiger S, Magi A, Bartalucci N, Raglianti V, Guzzi F, Maggi L, Annunziato F, Burger A, Lazzeri E, Anders HJ, Lasagni L, Romagnani P. Differentiation of crescent-forming kidney progenitor cells into podocytes attenuates severe glomerulonephritis in mice. Sci Transl Med 2022; 14:eabg3277. [PMID: 35947676 PMCID: PMC7614034 DOI: 10.1126/scitranslmed.abg3277] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Crescentic glomerulonephritis is characterized by vascular necrosis and parietal epithelial cell hyperplasia in the space surrounding the glomerulus, resulting in the formation of crescents. Little is known about the molecular mechanisms driving this process. Inducing crescentic glomerulonephritis in two Pax2Cre reporter mouse models revealed that crescents derive from clonal expansion of single immature parietal epithelial cells. Preemptive and delayed histone deacetylase inhibition with panobinostat, a drug used to treat hematopoietic stem cell disorders, attenuated crescentic glomerulonephritis with recovery of kidney function in the two mouse models. Three-dimensional confocal microscopy and stimulated emission depletion superresolution imaging of mouse glomeruli showed that, in addition to exerting an anti-inflammatory and immunosuppressive effect, panobinostat induced differentiation of an immature hyperplastic parietal epithelial cell subset into podocytes, thereby restoring the glomerular filtration barrier. Single-cell RNA sequencing of human renal progenitor cells in vitro identified an immature stratifin-positive cell subset and revealed that expansion of this stratifin-expressing progenitor cell subset was associated with a poor outcome in human crescentic glomerulonephritis. Treatment of human parietal epithelial cells in vitro with panobinostat attenuated stratifin expression in renal progenitor cells, reduced their proliferation, and promoted their differentiation into podocytes. These results offer mechanistic insights into the formation of glomerular crescents and demonstrate that selective targeting of renal progenitor cells can attenuate crescent formation and the deterioration of kidney function in crescentic glomerulonephritis in mice.
Collapse
Affiliation(s)
- Maria Elena Melica
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Giulia Antonelli
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Roberto Semeraro
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Maria Lucia Angelotti
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Gianmarco Lugli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Samuela Landini
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Fiammetta Ravaglia
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Gilda La Regina
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Carolina Conte
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Letizia De Chiara
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Anna Julie Peired
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Benedetta Mazzinghi
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Marta Donati
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Alice Molli
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Stefanie Steiger
- Division of Nephrology, Medizinische Klinik and Poliklinik IV, Klinikum der LMU München, Munich 80336, Germany
| | - Alberto Magi
- Department of Information Engineering, University of Florence, Florence, Italy
| | - Niccolò Bartalucci
- Department of Experimental and Clinical Medicine, CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, AOUC, University of Florence, Florence 50139, Italy
| | - Valentina Raglianti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Francesco Guzzi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alexa Burger
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Elena Lazzeri
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Hans-Joachim Anders
- Division of Nephrology, Medizinische Klinik and Poliklinik IV, Klinikum der LMU München, Munich 80336, Germany
| | - Laura Lasagni
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Corresponding authors. and
| | - Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy,Corresponding authors. and
| |
Collapse
|
17
|
Gilchrist AE, Harley BA. Engineered Tissue Models to Replicate Dynamic Interactions within the Hematopoietic Stem Cell Niche. Adv Healthc Mater 2022; 11:e2102130. [PMID: 34936239 PMCID: PMC8986554 DOI: 10.1002/adhm.202102130] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/19/2021] [Indexed: 12/19/2022]
Abstract
Hematopoietic stem cells are the progenitors of the blood and immune system and represent the most widely used regenerative therapy. However, their rarity and limited donor base necessitate the design of ex vivo systems that support HSC expansion without the loss of long-term stem cell activity. This review describes recent advances in biomaterials systems to replicate features of the hematopoietic niche. Inspired by the native bone marrow, these instructive biomaterials provide stimuli and cues from cocultured niche-associated cells to support HSC encapsulation and expansion. Engineered systems increasingly enable study of the dynamic nature of the matrix and biomolecular environment as well as the role of cell-cell signaling (e.g., autocrine feedback vs paracrine signaling between dissimilar cells). The inherent coupling of material properties, biotransport of cell-secreted factors, and cell-mediated remodeling motivate dynamic biomaterial systems as well as characterization and modeling tools capable of evaluating a temporally evolving tissue microenvironment. Recent advances in HSC identification and tracking, model-based experimental design, and single-cell culture platforms facilitate the study of the effect of constellations of matrix, cell, and soluble factor signals on HSC fate. While inspired by the HSC niche, these tools are amenable to the broader stem cell engineering community.
Collapse
Affiliation(s)
- Aidan E. Gilchrist
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Brendan A.C. Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
18
|
Chen Z, Guo Q, Song G, Hou Y. Molecular regulation of hematopoietic stem cell quiescence. Cell Mol Life Sci 2022; 79:218. [PMID: 35357574 PMCID: PMC11072845 DOI: 10.1007/s00018-022-04200-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 02/07/2022] [Accepted: 02/07/2022] [Indexed: 12/19/2022]
Abstract
Hematopoietic stem cells (HSCs) are primarily dormant in a cell-cycle quiescence state to preserve their self-renewal capacity and long-term maintenance, which is essential for the homeostasis of hematopoietic system. Dysregulation of quiescence causes HSC dysfunction and may result in aberrant hematopoiesis (e.g., myelodysplastic syndrome and bone marrow failure syndromes) and leukemia transformation. Accumulating evidence indicates that both intrinsic molecular networks and extrinsic signals regulate HSC quiescence, including cell-cycle regulators, transcription factors, epigenetic factors, and niche factors. Further, the transition between quiescence and activation of HSCs is a continuous developmental path driven by cell metabolism (e.g., protein synthesis, glycolysis, oxidative phosphorylation, and autophagy). Elucidating the complex regulatory networks of HSC quiescence will expand the knowledge of HSC hemostasis and benefit for clinical HSC use. Here, we review the current understanding and progression on the molecular and metabolic regulation of HSC quiescence, providing a more complete picture regarding the mechanisms of HSC quiescence maintenance.
Collapse
Affiliation(s)
- Zhe Chen
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Qian Guo
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Yu Hou
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, China.
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
19
|
The extracellular matrix of hematopoietic stem cell niches. Adv Drug Deliv Rev 2022; 181:114069. [PMID: 34838648 PMCID: PMC8860232 DOI: 10.1016/j.addr.2021.114069] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 12/21/2022]
Abstract
Comprehensive overview of different classes of ECM molecules in the HSC niche. Overview of current knowledge on role of biophysics of the HSC niche. Description of approaches to create artificial stem cell niches for several application. Importance of considering ECM in drug development and testing.
Hematopoietic stem cells (HSCs) are the life-long source of all types of blood cells. Their function is controlled by their direct microenvironment, the HSC niche in the bone marrow. Although the importance of the extracellular matrix (ECM) in the niche by orchestrating niche architecture and cellular function is widely acknowledged, it is still underexplored. In this review, we provide a comprehensive overview of the ECM in HSC niches. For this purpose, we first briefly outline HSC niche biology and then review the role of the different classes of ECM molecules in the niche one by one and how they are perceived by cells. Matrix remodeling and the emerging importance of biophysics in HSC niche function are discussed. Finally, the application of the current knowledge of ECM in the niche in form of artificial HSC niches for HSC expansion or targeted differentiation as well as drug testing is reviewed.
Collapse
|
20
|
Abnormal B-cell development in TIMP-deficient bone marrow. Blood Adv 2021; 5:3960-3974. [PMID: 34500457 PMCID: PMC8945646 DOI: 10.1182/bloodadvances.2020004101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 06/09/2021] [Indexed: 11/20/2022] Open
Abstract
Bone marrow (BM) is the primary site of hematopoiesis and is responsible for a lifelong supply of all blood cell lineages. The process of hematopoiesis follows key intrinsic programs that also integrate instructive signals from the BM niche. First identified as an erythropoietin-potentiating factor, the tissue inhibitor of metalloproteinase (TIMP) protein family has expanded to 4 members and has widely come to be viewed as a classical regulator of tissue homeostasis. By virtue of metalloprotease inhibition, TIMPs not only regulate extracellular matrix turnover but also control growth factor bioavailability. The 4 mammalian TIMPs possess overlapping enzyme-inhibition profiles and have never been studied for their cumulative role in hematopoiesis. Here, we show that TIMPs are critical for postnatal B lymphopoiesis in the BM. TIMP-deficient mice have defective B-cell development arising at the pro-B-cell stage. Expression analysis of TIMPless hematopoietic cell subsets pointed to an altered B-cell program in the Lineage-Sca-1+c-Kit+ (LSK) cell fraction. Serial and competitive BM transplants identified a defect in TIMP-deficient hematopoietic stem and progenitor cells for B lymphopoiesis. In parallel, reverse BM transplants uncovered the extrinsic role of stromal TIMPs in pro- and pre-B-cell development. TIMP deficiency disrupted CXCL12 localization to LepR+ cells, and increased soluble CXCL12 within the BM niche. It also compromised the number and morphology of LepR+ cells. These data provide new evidence that TIMPs control the cellular and biochemical makeup of the BM niche and influence the LSK transcriptional program required for optimal B lymphopoiesis.
Collapse
|
21
|
Habič A, Novak M, Majc B, Lah Turnšek T, Breznik B. Proteases Regulate Cancer Stem Cell Properties and Remodel Their Microenvironment. J Histochem Cytochem 2021; 69:775-794. [PMID: 34310223 DOI: 10.1369/00221554211035192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Proteolytic activity is perturbed in tumors and their microenvironment, and proteases also affect cancer stem cells (CSCs). CSCs are the therapy-resistant subpopulation of cancer cells with tumor-initiating capacity that reside in specialized tumor microenvironment niches. In this review, we briefly summarize the significance of proteases in regulating CSC activities with a focus on brain tumor glioblastoma. A plethora of proteases and their inhibitors participate in CSC invasiveness and affect intercellular interactions, enhancing CSC immune, irradiation, and chemotherapy resilience. Apart from their role in degrading the extracellular matrix enabling CSC migration in and out of their niches, we review the ability of proteases to modulate CSC properties, which prevents their elimination. When designing protease-oriented therapies, the multifaceted roles of proteases should be thoroughly investigated.
Collapse
Affiliation(s)
- Anamarija Habič
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia.,The Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Metka Novak
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Bernarda Majc
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia.,The Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Tamara Lah Turnšek
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia.,The Jožef Stefan International Postgraduate School, Ljubljana, Slovenia.,Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Barbara Breznik
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| |
Collapse
|
22
|
O'Reilly E, Zeinabad HA, Szegezdi E. Hematopoietic versus leukemic stem cell quiescence: Challenges and therapeutic opportunities. Blood Rev 2021; 50:100850. [PMID: 34049731 DOI: 10.1016/j.blre.2021.100850] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/22/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022]
Abstract
Hematopoietic stem cells (HSC) are responsible for the production of mature blood cells. To ensure that the HSC pool does not get exhausted over the lifetime of an individual, most HSCs are in a state of quiescence with only a small proportion of HSCs dividing at any one time. HSC quiescence is carefully controlled by both intrinsic and extrinsic, niche-driven mechanisms. In acute myeloid leukemia (AML), the leukemic cells overtake the hematopoietic bone marrow niche where they acquire a quiescent state. These dormant AML cells are resistant to chemotherapeutics. Because they can re-establish the disease after therapy, they are often termed as quiescent leukemic stem cells (LSC) or leukemia-initiating cells. While advancements are being made to target particular driver mutations in AML, there is less focus on how to tackle the drug resistance of quiescent LSCs. This review summarises the current knowledge on the biochemical characteristics of quiescent HSCs and LSCs, the intracellular signaling pathways and the niche-driven mechanisms that control quiescence and the key differences between HSC- and LSC-quiescence that may be exploited for therapy.
Collapse
Affiliation(s)
- Eimear O'Reilly
- Apoptosis Research Centre, Department of Biochemistry, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Hojjat Alizadeh Zeinabad
- Apoptosis Research Centre, Department of Biochemistry, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Eva Szegezdi
- Apoptosis Research Centre, Department of Biochemistry, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
23
|
Hausinger R, Hackl M, Jardon Alvarez A, Kehr M, Romero Marquez S, Hettler F, Kehr C, Grziwok S, Schreck C, Peschel C, Istvánffy R, Oostendorp RAJ. Cathepsin K maintains the compartment of bone marrow T lymphocytes in vivo. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:521-532. [PMID: 33592138 PMCID: PMC8127559 DOI: 10.1002/iid3.412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 01/03/2023]
Abstract
In this study, we investigated the influence of the loss of cathepsin K (Ctsk) gene on the hematopoietic system in vitro and in vivo. We found that cultures with lineage- SCA1+ KIT+ (LSK) cells on Ctsk deficient stromal cells display reduced colony formation and proliferation, with increased differentiation, giving rise to repopulating cells with reduced ability to repopulate the donor LSKs and T cell compartments in the bone marrow (BM). Subsequent in vivo experiments showed impairment of lymphocyte numbers, but, gross effects on early hematopoiesis or myelopoiesis were not found. Most consistently in in vivo experimental settings, we found a significant reduction of (donor) T cell numbers in the BM. Lymphocyte deregulation is also found in transplantation experiments, which revealed that Ctsk is required for optimal regeneration of small populations of T cells, particularly in the BM, but also of thymic B cells. Interestingly, cell nonautonomous Ctsk regulates both B and T cell numbers, but T cell numbers in the BM require an additional autonomous Ctsk-dependent process. Thus, we show that Ctsk is required for the maintenance of hematopoietic stem cells in vitro, but in vivo, Ctsk deficiency most strongly affects lymphocyte homeostasis, particularly of T cells in the BM.
Collapse
Affiliation(s)
- Renate Hausinger
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III - Hematology and Oncology, Laboratory of Stem Cell Physiology, Munich, Germany
| | - Marianne Hackl
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III - Hematology and Oncology, Laboratory of Stem Cell Physiology, Munich, Germany
| | - Ana Jardon Alvarez
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III - Hematology and Oncology, Laboratory of Stem Cell Physiology, Munich, Germany
| | - Miriam Kehr
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III - Hematology and Oncology, Laboratory of Stem Cell Physiology, Munich, Germany
| | - Sandra Romero Marquez
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III - Hematology and Oncology, Laboratory of Stem Cell Physiology, Munich, Germany
| | - Franziska Hettler
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III - Hematology and Oncology, Laboratory of Stem Cell Physiology, Munich, Germany
| | - Christian Kehr
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III - Hematology and Oncology, Laboratory of Stem Cell Physiology, Munich, Germany
| | - Sandra Grziwok
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III - Hematology and Oncology, Laboratory of Stem Cell Physiology, Munich, Germany
| | - Christina Schreck
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III - Hematology and Oncology, Laboratory of Stem Cell Physiology, Munich, Germany
| | - Christian Peschel
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III - Hematology and Oncology, Laboratory of Stem Cell Physiology, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Rouzanna Istvánffy
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III - Hematology and Oncology, Laboratory of Stem Cell Physiology, Munich, Germany
| | - Robert A J Oostendorp
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III - Hematology and Oncology, Laboratory of Stem Cell Physiology, Munich, Germany
| |
Collapse
|
24
|
Aasebø E, Birkeland E, Selheim F, Berven F, Brenner AK, Bruserud Ø. The Extracellular Bone Marrow Microenvironment-A Proteomic Comparison of Constitutive Protein Release by In Vitro Cultured Osteoblasts and Mesenchymal Stem Cells. Cancers (Basel) 2020; 13:cancers13010062. [PMID: 33379263 PMCID: PMC7795818 DOI: 10.3390/cancers13010062] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Normal blood cells are formed in the bone marrow by a process called hematopoiesis. This process is supported by a network of non-hematopoietic cells including connective tissue cells, blood vessel cells and bone-forming cells. However, these cells can also support the growth of cancer cells, i.e., hematological malignancies (e.g., leukemias) and cancers that arise in another organ and spread to the bone marrow. Two of these cancer-supporting normal cells are bone-forming osteoblasts and a subset of connective tissue cells called mesenchymal stem cells. One mechanism for their cancer support is the release of proteins that support cancer cell proliferation and progression of the cancer disease. Our present study shows that both these normal cells release a wide range of proteins that support cancer cells, and inhibition of this protein-mediated cancer support may become a new strategy for cancer treatment. Abstract Mesenchymal stem cells (MSCs) and osteoblasts are bone marrow stromal cells that contribute to the formation of stem cell niches and support normal hematopoiesis, leukemogenesis and development of metastases from distant cancers. This support is mediated through cell–cell contact, release of soluble mediators and formation of extracellular matrix. By using a proteomic approach, we characterized the protein release by in vitro cultured human MSCs (10 donors) and osteoblasts (nine donors). We identified 1379 molecules released by these cells, including 340 proteins belonging to the GO-term Extracellular matrix. Both cell types released a wide range of functionally heterogeneous proteins including extracellular matrix molecules (especially collagens), several enzymes and especially proteases, cytokines and soluble adhesion molecules, but also several intracellular molecules including chaperones, cytoplasmic mediators, histones and non-histone nuclear molecules. The levels of most proteins did not differ between MSCs and osteoblasts, but 82 proteins were more abundant for MSC (especially extracellular matrix proteins and proteases) and 36 proteins more abundant for osteoblasts. Finally, a large number of exosomal proteins were identified. To conclude, MSCs and osteoblasts show extracellular release of a wide range of functionally diverse proteins, including several extracellular matrix molecules known to support cancer progression (e.g., metastases from distant tumors, increased relapse risk for hematological malignancies), and the large number of identified exosomal proteins suggests that exocytosis is an important mechanism of protein release.
Collapse
Affiliation(s)
- Elise Aasebø
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway; (E.A.); (A.K.B.)
| | - Even Birkeland
- The Proteomics Facility of the University of Bergen (PROBE), University of Bergen, N-5021 Bergen, Norway; (E.B.); (F.S.); (F.B.)
| | - Frode Selheim
- The Proteomics Facility of the University of Bergen (PROBE), University of Bergen, N-5021 Bergen, Norway; (E.B.); (F.S.); (F.B.)
| | - Frode Berven
- The Proteomics Facility of the University of Bergen (PROBE), University of Bergen, N-5021 Bergen, Norway; (E.B.); (F.S.); (F.B.)
| | - Annette K. Brenner
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway; (E.A.); (A.K.B.)
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway; (E.A.); (A.K.B.)
- Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway
- Correspondence: or ; Tel.: +47-5597-2997
| |
Collapse
|
25
|
Mishra S, Rajput C, Singh MP. Cypermethrin Induces the Activation of Rat Primary Microglia and Expression of Inflammatory Proteins. J Mol Neurosci 2020; 71:1275-1283. [PMID: 33230707 DOI: 10.1007/s12031-020-01753-y] [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: 07/13/2020] [Accepted: 11/08/2020] [Indexed: 10/22/2022]
Abstract
Cypermethrin activates microglia, which is found to be decisive in neurodegeneration in the experimental rats. While the involvement of microglial activation in toxicant-induced neurodegeneration is reported, the effect of low concentration of cypermethrin on the expression of inflammatory proteins from the rat primary microglia is not yet properly understood. The study intended to delineate the effect of low concentration of cypermethrin on the expression and release of proteins from the microglia. Rat primary microglial cells were treated with cypermethrin to check the expression of inflammatory proteins. Cypermethrin-treated microglia conditioned media and cells were collected to measure the expression and release of inflammatory proteins. Cypermethrin augmented the protein kinase C-δ (PKC-δ), inducible nitric oxide synthase (iNOS), phosphorylated mitogen-activated protein kinase (MAPK) p38 and p42/44, matrix metalloproteinase (MMP)-3, and MMP-9 levels in the cell lysate and tumour necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) levels in the microglia conditioned media. Pre-treatment with minocycline, a microglial activation inhibitor or rottlerin, a PKC-δ inhibitor, notably reduced the release of TNF-α in the conditioned media and expression of iNOS protein in the microglia. Minocycline reduced the expression of PKC-δ, phosphorylated p38 and p42/44 MAPKs, MMP-3, and MMP-9 proteins in the microglia. While cypermethrin-treated conditioned media induced the toxicity in the rat primary neurons, minocycline or rottlerin reduced the cypermethrin treated microglia conditioned media-induced toxicity. The outcomes of the present study suggest that cypermethrin activates microglia and releases TNF-α and IL-1β as well as up-regulates the expression of PKC-δ, iNOS, phosphorylated p38 and p42/44 MAPKs, MMP-3, and MMP-9 proteins, which could contribute to neurodegeneration.
Collapse
Affiliation(s)
- Saumya Mishra
- Toxicogenomics and Predictive Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201 002, Uttar Pradesh, India
| | - Charul Rajput
- Toxicogenomics and Predictive Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201 002, Uttar Pradesh, India
| | - Mahendra Pratap Singh
- Toxicogenomics and Predictive Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201 002, Uttar Pradesh, India.
| |
Collapse
|
26
|
Chen ZH, Luo XC, Yu CR, Huang L. Matrix metalloprotease-mediated cleavage of neural glial-related cell adhesion molecules activates quiescent olfactory stem cells via EGFR. Mol Cell Neurosci 2020; 108:103552. [PMID: 32918999 DOI: 10.1016/j.mcn.2020.103552] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/23/2020] [Accepted: 09/06/2020] [Indexed: 12/14/2022] Open
Abstract
Quiescent stem cells have been found in multiple adult organs, and activation of these stem cells is critical to the restoration of damaged tissues in response to injury or stress. Existing evidence suggests that extrinsic cues from the extracellular matrix or supporting cells of various stem cell niches may interact with intrinsic components to initiate stem cell differentiation, but the molecular and cellular mechanisms regulating their activation are not fully understood. In the present study, we find that olfactory horizontal basal cells (HBCs) are stimulated by neural glial-related cell adhesion molecules (NrCAMs). NrCAM activation requires matrix metalloproteases (MMPs) and epidermal growth factor receptors (EGFRs). Inhibiting MMP activity or EGFR activation not only blocks HBC proliferation in the cultured olfactory organoids, but also severely suppresses HBC proliferation in the olfactory epithelium following methimazole-induced injury, resulting in a delay of olfactory mucosa reconstitution and functional recovery of the injured mice. Both NrCAMs and EGFR are expressed by the HBCs and their expression increases upon injury. Our data indicate that MMP-mediated cleavage of NrCAMs serves as an autocrine or paracrine signal that activates EGFRs on HBCs to trigger HBC proliferation and differentiation to reconstruct the entire olfactory epithelium following injury.
Collapse
Affiliation(s)
- Zhen-Huang Chen
- Institute of Cellular and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiao-Cui Luo
- Institute of Cellular and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - C Ron Yu
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Liquan Huang
- Institute of Cellular and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, Zhejiang 310027, China; Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
27
|
Kumar P, Siripini S, Sreedhar AS. The matrix metalloproteinase 7 (MMP7) links Hsp90 chaperone with acquired drug resistance and tumor metastasis. Cancer Rep (Hoboken) 2020; 5:e1261. [PMID: 32761892 PMCID: PMC9780424 DOI: 10.1002/cnr2.1261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/12/2020] [Accepted: 06/01/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Cancer emergence is associated with a series of cellular transformations that include acquired drug resistance followed by tumor metastasis. Matrix metalloproteinases (MMPs) and Hsp90 chaperone are implicated in tumor progression, however, they are not studied in the context of drug resistance. AIMS In the present study, we aimed at understanding the cross-talk between acquired drug resistance and tumor progression, linking MMP7 and Hsp90. METHODS AND RESULTS We have developed an in vitro model system for acquired drug resistance and studied the correlation between MMP7 and Hsp90. We demonstrate that enhanced drug efflux activity correlates with the induced expression and activity of MMP7, and enhanced metastatic potential of cells, however, in Hsp90-dependent manner. The MMP7 overexpression alone could enhance the drug efflux activity marginally, and metastasis significantly. However, challenging these cells with 17AAG has significantly increased the drug efflux activity and, in contrast, decreased the metastatic potential. Evaluating our in vitro findings in mice xenografts revealed that MMP7 overexpression facilitates altered homing properties. However, these cells, in response to 17AAG treatment, exhibited increased localized tumor growth but decreased tumor metastasis. CONCLUSION We demonstrated a cross-talk between Hsp90 and MMP7 in regulating the acquired drug resistance and tumor progression. Our findings provide novel insights on targeting drug resistant-tumors.
Collapse
Affiliation(s)
- Pankaj Kumar
- CSIR‐Centre for Cellular and Molecular BiologyHyderabadIndia
| | | | | |
Collapse
|
28
|
Calf thymus polypeptide improved hematopoiesis via regulating colony-stimulating factors in BALB/c mice with hematopoietic dysfunction. Int J Biol Macromol 2020; 156:204-216. [PMID: 32156537 DOI: 10.1016/j.ijbiomac.2020.03.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/31/2022]
Abstract
Calf thymus polypeptide (CTP) is prepared from calf thymus. It has a molecular mass of <10 kilodalton (kDa) and contains 17 types of amino acids. This study investigated the hematopoietic function-improvement effect of CTP in CHRF, K562, and bone marrow mononuclear cells; mice with immunosuppression; and with hematopoietic dysfunction. In mice with immunosuppression, CTP enhanced the cytotoxic activity of natural killer cells and the proliferation of lymphocytes and regulated the levels of immunoglobulins. It also enhanced the proliferation and differentiation of CHRF and K562 cells by upregulating the expression of proliferation- and differentiation-related proteins. In mice with hematopoietic dysfunction, CTP restored white blood cell, neutrophil, and hemoglobin proportions in the peripheral blood and enhanced the levels of B lymphocytes and hematopoietic stem cells and progenitor cells in the bone marrow. CTP effectively regulated the levels of hematopoiesis-related cytokines, such as granulocyte colony-stimulating factor (G-CSF), macrophage colony-stimulating factor (M-CSF), interleukin 2, and interferons-γ, and enhanced the expression of hematopoiesis-related proteins in both primary bone marrow cells and mice with hematopoietic dysfunction. These results indicate that CTP has hematopoietic function-improvement effect and this effect may be related to the modulation of colony-stimulating factors (CSFs) and related signaling pathways.
Collapse
|