1
|
Feng J, Zhang M, Ren H, Ren Y, Hao Z, Bian S, Cui J, Li S, Xu J, Daniel MM, Ren F, Xu Z, Tan Y, Chen X, Zhang Y, Chang J, Wang H. Human umbilical cord mesenchymal stem cells improve bone marrow hematopoiesis through regulation of bone marrow adipose tissue. Mol Cell Biochem 2025; 480:3033-3049. [PMID: 39613944 PMCID: PMC12048464 DOI: 10.1007/s11010-024-05156-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 11/02/2024] [Indexed: 12/01/2024]
Abstract
Bone marrow adipose tissue (BMAT) exhibits a multitude of biological functionalities and influences hematopoiesis. The adiposity status of the bone marrow may play a role in the decline of hematopoietic function. Mesenchymal stem cells (MSCs) constitute crucial regulators within the bone marrow microenvironment; however, their precise role in modulating BMAT and the subsequent implications for hematopoiesis remain poorly understood. We conducted in vivo studies to observe the effects of human umbilical cord mesenchymal stem cells (hucMSCs) on BMAT accumulation and restoration of hematopoietic function in mice with drug-induced hematopoietic impairment. Concurrently, in vitro co-culture experiments were used to investigate the impact of hucMSCs on preadipocytes and mature adipocytes, and the potential subsequent consequences for hematopoietic cells. Moreover, we explored the potential mechanisms underlying these interactions. Our findings reveal that hucMSCs concomitantly mitigate BMAT accumulation and facilitate the recovery of hematopoietic function in mouse models with drug-induced hematopoietic impairment. In vitro, hucMSCs potentially impede adipogenic differentiation of 3T3-L1 preadipocytes through interference with the JAK2/STAT3 signaling pathway and affect the functionality of mature adipocytes, thus mitigating the detrimental effects of adipocytes on hematopoietic stem cells (HSCs). Furthermore, we demonstrate that hucMSCs may protect hematopoietic cells from adipocyte-induced damage by protecting antioxidative mechanisms. These results suggest that hucMSCs exhibit an inhibitory effect on the excessive expansion of adipose tissue and modulate adipose tissue function, which may potentially contribute to the regulation of the bone marrow microenvironment and favorably influence hematopoietic function improvement.
Collapse
Affiliation(s)
- Jingyi Feng
- Institute of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, People's Republic of China
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Miao Zhang
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Huanying Ren
- Institute of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Yan Ren
- Institute of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Zhuanghui Hao
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Sicheng Bian
- Department of Medicine, The MetroHealth System, Case Western Reserve University, Cleveland, OH, 44109, USA
| | - Jiangxia Cui
- Institute of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Shuo Li
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Jing Xu
- Department of Medical Cell Biology and Genetics, Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Muteb Muyey Daniel
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Fanggang Ren
- Institute of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Zhifang Xu
- Institute of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Yanhong Tan
- Institute of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Xiuhua Chen
- Institute of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Yaofang Zhang
- Institute of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Jianmei Chang
- Institute of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Hongwei Wang
- Institute of Hematology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, People's Republic of China.
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, People's Republic of China.
| |
Collapse
|
2
|
Liu X, Zhang H, Yan J, Ye P, Wang Y, Zhang N, Tian Z, Liu B, Yang H. Purine metabolism in bone marrow microenvironment inhibits hematopoietic stem cell differentiation under microgravity. Stem Cell Res Ther 2025; 16:115. [PMID: 40038750 PMCID: PMC11881365 DOI: 10.1186/s13287-025-04213-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/30/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Spaceflight and microgravity environments have been shown to cause significant health impairments, including bone loss, immune dysfunction, and hematopoietic disorders. Hematopoietic stem cells (HSCs), as progenitors of the hematopoietic system, are critical for the continuous renewal and regulation of immune cells. Therefore, elucidating the regulatory mechanisms governing HSC fate and differentiation in microgravity environments is of paramount importance. METHODS In this study, hindlimb unloading (HU) was employed in mice to simulate microgravity conditions. After 28 days of HU, cells were isolated for analysis. Flow cytometry and colony-forming assays were utilized to assess changes in HSC proliferation and differentiation. Additionally, transcriptomic and untargeted metabolomic sequencing were performed to elucidate alterations in the metabolic pathways of the bone marrow microenvironment and their molecular regulatory effects on HSCs fate. RESULTS Our findings revealed that 28 days of HU impaired hematopoietic function, leading to multi-organ damage and hematological disorders. The simulated microgravity environment significantly increased the HSCs population in the bone marrow, particularly within the long-term and short-term subtypes, while severely compromising the differentiation capacity of hematopoietic stem/progenitor cells. Transcriptomic analysis of HSCs, combined with metabolomic profiling of bone marrow supernatants, identified 1,631 differentially expressed genes and 58 metabolites with altered abundance. Gene set enrichment analysis indicated that HU suppressed key pathways, including hematopoietic cell lineage and MAPK signaling. Furthermore, integrated analyses revealed that metabolites affected by HU, particularly hypoxanthine enriched in the purine metabolism pathway, were closely associated with hematopoietic cell lineage and MAPK signaling pathways. Molecular docking simulations and in vitro experiments confirmed that hypoxanthine interacts directly with core molecules within these pathways, influencing their expression. CONCLUSIONS These findings demonstrate that hypoxanthine in the bone marrow supernatant acts as a signaling mediator under microgravity, influencing HSCs fate by modulating hematopoietic cell lineage and MAPK signaling pathways. This study offers novel insights into the impact of microgravity on HSC fate and gene expression, underscoring the pivotal role of bone marrow microenvironmental metabolic changes in regulating key signaling pathways that determine hematopoietic destiny.
Collapse
Affiliation(s)
- Xiru Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, China
- Research Center of Special Environmental Biomechanics and Medical Engineering, Northwestern Polytechnical University, Xi'an, China
| | - Hao Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, China
- Research Center of Special Environmental Biomechanics and Medical Engineering, Northwestern Polytechnical University, Xi'an, China
| | - Jinxiao Yan
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, China
- Research Center of Special Environmental Biomechanics and Medical Engineering, Northwestern Polytechnical University, Xi'an, China
| | - Penghui Ye
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, China
- Research Center of Special Environmental Biomechanics and Medical Engineering, Northwestern Polytechnical University, Xi'an, China
| | - Yanran Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, China
- Research Center of Special Environmental Biomechanics and Medical Engineering, Northwestern Polytechnical University, Xi'an, China
| | - Nu Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, China
- Research Center of Special Environmental Biomechanics and Medical Engineering, Northwestern Polytechnical University, Xi'an, China
| | - Zhenhao Tian
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, China
- Research Center of Special Environmental Biomechanics and Medical Engineering, Northwestern Polytechnical University, Xi'an, China
| | - Bin Liu
- Department of Infectious Diseases, Characteristic Medical Center of Chinese People's Armed Police Forces, Tianjin, China.
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.
- Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Xi'an, China.
- Research Center of Special Environmental Biomechanics and Medical Engineering, Northwestern Polytechnical University, Xi'an, China.
| |
Collapse
|
3
|
Albiero M, Baragetti A. Exploring neutrophils as therapeutic targets in cardiometabolic diseases. Trends Pharmacol Sci 2025; 46:102-116. [PMID: 39855946 DOI: 10.1016/j.tips.2024.12.003] [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: 05/24/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 01/27/2025]
Abstract
Current therapies for diabetes and atherosclerotic cardiovascular diseases (ACVDs) mainly target metabolic risk factors, but often fall short in addressing systemic inflammation, a key driver of disease onset and progression. Advances in our understanding of the biology of neutrophils, the cells that are principally involved in inflammatory situations, have highlighted their pivotal role in cardiometabolic diseases. Yet, neutrophils can reprogram their immune-metabolic functions based on the energetic substrates available, thus influencing both tissue homeostasis and the resolution of inflammation. In this review, we examine the effects of canonical therapies for cardiometabolic diseases on the key molecular pathways through which neutrophils respond to inflammatory stimuli. In addition, we explore potential synergies between these established therapeutic approaches and the anti-inflammatory therapies being evaluated for repurposing in the treatment of cardiometabolic diseases.
Collapse
Affiliation(s)
- Mattia Albiero
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, Padua, Italy; Regional Center for the Cellular Therapy of Diabetes, University Hospital of Padova, Padua, Italy; Veneto Institute of Molecular Medicine, Laboratory of Experimental Diabetology, Padua, Italy.
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
4
|
Liu H, Liu L, Rosen CJ. Bone Marrow Adipocytes as Novel Regulators of Metabolic Homeostasis: Clinical Consequences of Bone Marrow Adiposity. Curr Obes Rep 2025; 14:9. [PMID: 39808256 DOI: 10.1007/s13679-024-00594-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE OF REVIEW Bone marrow adipose tissue is a distinctive fat depot located within the skeleton, with the potential to influence both local and systemic metabolic processes. Although significant strides have been made in understanding bone marrow adipose tissue over the past decade, many questions remain regarding their precise lineage and functional roles. RECENT FINDINGS Recent studies have highlighted bone marrow adipose tissue's involvement in continuous cross-talk with other organs and systems, exerting both endocrine and paracrine functions that play a crucial role in metabolic homeostasis, skeletal remodeling, hematopoiesis, and the progression of bone metastases. The advancement of imaging techniques, particularly cross-sectional imaging, has profoundly expanded our understanding of the complexities beyond the traditional view of bone marrow adipose tissue as an inert depot. Notably, marrow adipocytes are anatomically and functionally distinct from brown, beige, and classic white adipocytes. Emerging evidence suggests that bone marrow adipocytes, bone marrow adipose tissue originate from the differentiation of bone marrow mesenchymal stromal cells; however, they appear to be a heterogeneous population with varying metabolic profiles, lipid compositions, secretory properties, and functional responses depending on their specific location within the bone marrow. This review provides an up-to-date synthesis of current knowledge on bone marrow adipocytes, emphasizing the relationships between bone marrow adipogenesis and factors such as aging, osteoporosis, obesity, and bone marrow tumors or metastases, thereby elucidating the mechanisms underlying musculoskeletal pathophysiology.
Collapse
Affiliation(s)
- Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology &, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Linyi Liu
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA.
| |
Collapse
|
5
|
Trivanović D, Vujačić M, Arsić A, Kukolj T, Rajković M, Bogosavljević N, Baščarević Z, Maljković Ružičić M, Kovačević J, Jauković A. Skeletal Site-Specific Lipid Profile and Hematopoietic Progenitors of Bone Marrow Adipose Tissue in Patients Undergoing Primary Hip Arthroplasty. Metabolites 2025; 15:16. [PMID: 39852359 PMCID: PMC11767117 DOI: 10.3390/metabo15010016] [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: 12/10/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Bone marrow adipose tissue (BMAT) has been described as an important biomechanic and lipotoxic factor with negative impacts on skeletal and hematopoietic system regeneration. BMAT undergoes metabolic and cellular adaptations with age and disease, being a source of potential biomarkers. However, there is no evidence on the lipid profile and cellularity at different skeletal locations in osteoarthritis patients undergoing primary hip arthroplasty. METHODS Acetabular and femoral bone marrow (BM) and gluteofemoral subcutaneous adipose tissue (gfSAT) were obtained from matched patients undergoing hip replacement surgery. BM, BMAT, and gfSAT were explored at the levels of total lipids, fatty acids, and cells by using thin-layerand gas chromatography, ex vivo cellular assays, and flow cytometry. RESULTS BMAT content was significantly higher in femoral than in acetabular BM. Total lipid analyses revealed significantly lower triglyceride content in femoral than in acetabular BMAT and gfSAT. Frequencies of saturated palmitic, myristic, and stearic acids were higher in femoral than in acetabular BMAT and gfSAT. The content of CD45+CD34+ cells within femoral BMAT was higher than in acetabular BMAT or gfSAT. This was associated with a higher incidence of total clonogenic hematopoietic progenitors and late erythroid colonies CFU-E in femoral BMAT when compared to acetabular BMAT, similar to their BM counterparts. CONCLUSIONS Collectively, our results indicate that the lipid profiles of hip bone and femoral BMAT impose significantly different microenvironments and distributions of cells with hematopoietic potential. These findings might bring forth new inputs for defining BMAT biology and setting novel directions in OA disease investigations.
Collapse
Affiliation(s)
- Drenka Trivanović
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (T.K.); (M.R.); (A.J.)
| | - Marko Vujačić
- Institute for Orthopedy Banjica, 11000 Belgrade, Serbia; (M.V.); (N.B.); (Z.B.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Aleksandra Arsić
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Tamara Kukolj
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (T.K.); (M.R.); (A.J.)
| | - Milica Rajković
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (T.K.); (M.R.); (A.J.)
| | - Nikola Bogosavljević
- Institute for Orthopedy Banjica, 11000 Belgrade, Serbia; (M.V.); (N.B.); (Z.B.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Zoran Baščarević
- Institute for Orthopedy Banjica, 11000 Belgrade, Serbia; (M.V.); (N.B.); (Z.B.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | | | - Jovana Kovačević
- Faculty of Mathematics, University of Belgrade, 11000 Belgrade, Serbia; (M.M.R.); (J.K.)
- Institute for Artificial Intelligence Research and Development of Serbia, 21000 Novi Sad, Serbia
| | - Aleksandra Jauković
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (T.K.); (M.R.); (A.J.)
| |
Collapse
|
6
|
Deng S, Zhang S, Shen T, Wang X, Gao Z, Zhang W, Dai K, Wang J, Liu C. Amphiphilic cytokine traps remodel marrow adipose tissue for hematopoietic microenvironment amelioration. Bioact Mater 2024; 42:226-240. [PMID: 39285915 PMCID: PMC11404087 DOI: 10.1016/j.bioactmat.2024.08.032] [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: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is extensively employed in the treatment of hematological malignancies but is markedly constrained by the paucity of hematopoietic stem/progenitor cells (HSPCs). Recent studies have found that marrow adipose tissue (MAT) acts on hematopoiesis through complicated mechanisms. Therefore, the osteo-organoids fabricated in vivo using biomaterials loaded with recombinant human bone morphogenetic protein 2 (rhBMP-2) have been used as models of MAT for our research. To obtain sufficient amounts of therapeutic HSPCs and healthy MAT, we have developed amphiphilic chitosan (AC)-gelatin as carriers of rhBMP-2 to the regulate type conversion of adipose tissue and trap hematopoietic growth factors. Unlike medicine interventions or cell therapies, the traps based on AC not only attenuate the occupancy of adipocytes within the hematopoietic microenvironment while preserving stem cell factor concentrations, but also improve marrow metabolism by promoting MAT browning. In conclusion, this approach increases the proportion of HSPCs in osteo-organoids, and optimizes the composition and metabolic status of MAT. These findings furnish an experimental basis for regulating hematopoiesis in vivo through materials that promote the development of autologous HSPCs. Additionally, this approach presents a theoretical model of rapid adipogenesis for the study of adipose-related pathologies and potential pharmacological targets.
Collapse
Affiliation(s)
- Shunshu Deng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Shuang Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Tong Shen
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Xuanlin Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Zehua Gao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Wenchao Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Kai Dai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Jing Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
7
|
Carey AE, Weeraratna AT. Entering the TiME machine: How age-related changes in the tumor immune microenvironment impact melanoma progression and therapy response. Pharmacol Ther 2024; 262:108698. [PMID: 39098769 DOI: 10.1016/j.pharmthera.2024.108698] [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: 02/29/2024] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024]
Abstract
Melanoma is the deadliest form of skin cancer in the United States, with its incidence rates rising in older populations. As the immune system undergoes age-related changes, these alterations can significantly influence tumor progression and the effectiveness of cancer treatments. Recent advancements in understanding immune checkpoint molecules have paved the way for the development of innovative immunotherapies targeting solid tumors. However, the aging tumor microenvironment can play a crucial role in modulating the response to these immunotherapeutic approaches. This review seeks to examine the intricate relationship between age-related changes in the immune system and their impact on the efficacy of immunotherapies, particularly in the context of melanoma. By exploring this complex interplay, we hope to elucidate potential strategies to optimize treatment outcomes for older patients with melanoma, and draw parallels to other cancers.
Collapse
Affiliation(s)
- Alexis E Carey
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ashani T Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
8
|
Parolini A, Da Dalt L, Norata GD, Baragetti A. Dietary fats as regulators of neutrophil plasticity: an update on molecular mechanisms. Curr Opin Clin Nutr Metab Care 2024; 27:434-442. [PMID: 39083430 PMCID: PMC11309349 DOI: 10.1097/mco.0000000000001055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
PURPOSE OF REVIEW Contemporary guidelines for the prevention of cardio-metabolic diseases focus on the control of dietary fat intake, because of their adverse metabolic effects. Moreover, fats alter innate immune defenses, by eliciting pro-inflammatory epigenetic mechanisms on the long-living hematopoietic cell progenitors which, in the bone marrow, mainly give rise to short-living neutrophils. Nevertheless, the heterogenicity of fats and the complexity of the biology of neutrophils pose challenges in the understanding on how this class of nutrients could contribute to the development of cardio-metabolic diseases via specific molecular mechanisms activating the inflammatory response. RECENT FINDINGS The knowledge on the biology of neutrophils is expanding and there are now different cellular networks orchestrating site-specific reprogramming of these cells to optimize the responses against pathogens. The innate immune competence of neutrophil is altered in response to high fat diet and contributes to the development of metabolic alterations, although the precise mechanisms are still poorly understood. SUMMARY Defining the different molecular mechanisms involved in the fat-neutrophil crosstalk will help to reconcile the sparse data about the interaction of dietary fats with neutrophils and to tailor strategies to target neutrophils in the context of cardio-metabolic diseases.
Collapse
Affiliation(s)
- Anna Parolini
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy
| | | | | | | |
Collapse
|
9
|
Zhang X, Tian L, Majumdar A, Scheller EL. Function and Regulation of Bone Marrow Adipose Tissue in Health and Disease: State of the Field and Clinical Considerations. Compr Physiol 2024; 14:5521-5579. [PMID: 39109972 PMCID: PMC11725182 DOI: 10.1002/cphy.c230016] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
Bone marrow adipose tissue (BMAT) is a metabolically and clinically relevant fat depot that exists within bone. Two subtypes of BMAT, regulated and constitutive, reside in hematopoietic-rich red marrow and fatty yellow marrow, respectively, and exhibit distinct characteristics compared to peripheral fat such as white and brown adipose tissues. Bone marrow adipocytes (BMAds) are evolutionally preserved in most vertebrates, start development after birth and expand throughout life, and originate from unique progenitor populations that control bone formation and hematopoiesis. Mature BMAds also interact closely with other cellular components of the bone marrow niche, serving as a nearby energy reservoir to support the skeletal system, a signaling hub that contributes to both local and systemic homeostasis, and a final fuel reserve for survival during starvation. Though BMAT and bone are often inversely correlated, more BMAT does not always mean less bone, and the prevention of BMAT expansion as a strategy to prevent bone loss remains questionable. BMAT adipogenesis and lipid metabolism are regulated by the nervous systems and a variety of circulating hormones. This contributes to the plasticity of BMAT, including BMAT expansion in common physiological or pathological conditions, and BMAT catabolism under certain extreme circumstances, which are often associated with malnutrition and/or systemic inflammation. Altogether, this article provides a comprehensive overview of the local and systemic functions of BMAT and discusses the regulation and plasticity of this unique adipose tissue depot in health and disease. © 2024 American Physiological Society. Compr Physiol 14:5521-5579, 2024.
Collapse
Affiliation(s)
- Xiao Zhang
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Linda Tian
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Anurag Majumdar
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Erica L. Scheller
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
- Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
10
|
Yang Y, Zhou X, Deng H, Chen L, Zhang X, Wu S, Song A, Liang F. The role of O-GlcNAcylation in bone metabolic diseases. Front Physiol 2024; 15:1416967. [PMID: 38915778 PMCID: PMC11194333 DOI: 10.3389/fphys.2024.1416967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/20/2024] [Indexed: 06/26/2024] Open
Abstract
O-GlcNAcylation, as a post-translational modification, can modulate cellular activities such as kinase activity, transcription-translation, protein degradation, and insulin signaling by affecting the function of the protein substrate, including cellular localization of proteins, protein stability, and protein/protein interactions. Accumulating evidence suggests that dysregulation of O-GlcNAcylation is associated with disease progression such as cancer, neurodegeneration, and diabetes. Recent studies suggest that O-GlcNAcylation is also involved in the regulation of osteoblast, osteoclast and chondrocyte differentiation, which is closely related to the initiation and development of bone metabolic diseases such as osteoporosis, arthritis and osteosarcoma. However, the potential mechanisms by which O-GlcNAcylation regulates bone metabolism are not fully understood. In this paper, the literature related to the regulation of bone metabolism by O-GlcNAcylation was summarized to provide new potential therapeutic strategies for the treatment of orthopedic diseases such as arthritis and osteoporosis.
Collapse
Affiliation(s)
- Yajing Yang
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Xuchang Zhou
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
- School of Medicine, Xiamen University, Xiamen, China
| | - HuiLi Deng
- School of Medicine, Xiamen University, Xiamen, China
| | - Li Chen
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| | - Xiaolin Zhang
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| | - Song Wu
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| | - Aiqun Song
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| | - Fengxia Liang
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| |
Collapse
|
11
|
Bandyopadhyay S, Duffy MP, Ahn KJ, Sussman JH, Pang M, Smith D, Duncan G, Zhang I, Huang J, Lin Y, Xiong B, Imtiaz T, Chen CH, Thadi A, Chen C, Xu J, Reichart M, Martinez Z, Diorio C, Chen C, Pillai V, Snaith O, Oldridge D, Bhattacharyya S, Maillard I, Carroll M, Nelson C, Qin L, Tan K. Mapping the cellular biogeography of human bone marrow niches using single-cell transcriptomics and proteomic imaging. Cell 2024; 187:3120-3140.e29. [PMID: 38714197 PMCID: PMC11162340 DOI: 10.1016/j.cell.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 02/02/2024] [Accepted: 04/12/2024] [Indexed: 05/09/2024]
Abstract
Non-hematopoietic cells are essential contributors to hematopoiesis. However, heterogeneity and spatial organization of these cells in human bone marrow remain largely uncharacterized. We used single-cell RNA sequencing (scRNA-seq) to profile 29,325 non-hematopoietic cells and discovered nine transcriptionally distinct subtypes. We simultaneously profiled 53,417 hematopoietic cells and predicted their interactions with non-hematopoietic subsets. We employed co-detection by indexing (CODEX) to spatially profile over 1.2 million cells. We integrated scRNA-seq and CODEX data to link predicted cellular signaling with spatial proximity. Our analysis revealed a hyperoxygenated arterio-endosteal neighborhood for early myelopoiesis, and an adipocytic localization for early hematopoietic stem and progenitor cells (HSPCs). We used our CODEX atlas to annotate new images and uncovered mesenchymal stromal cell (MSC) expansion and spatial neighborhoods co-enriched for leukemic blasts and MSCs in acute myeloid leukemia (AML) patient samples. This spatially resolved, multiomic atlas of human bone marrow provides a reference for investigation of cellular interactions that drive hematopoiesis.
Collapse
Affiliation(s)
- Shovik Bandyopadhyay
- Cellular and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Medical Scientist Training Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P Duffy
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kyung Jin Ahn
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jonathan H Sussman
- Medical Scientist Training Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Minxing Pang
- Applied Mathematics & Computational Science Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - David Smith
- Center for Single Cell Biology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gwendolyn Duncan
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Iris Zhang
- Department of Computer and Information Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Yulieh Lin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Barbara Xiong
- Medical Scientist Training Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tamjid Imtiaz
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Chia-Hui Chen
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anusha Thadi
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Changya Chen
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jason Xu
- Medical Scientist Training Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Melissa Reichart
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zachary Martinez
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Caroline Diorio
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery/Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vinodh Pillai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Oraine Snaith
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Derek Oldridge
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Siddharth Bhattacharyya
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ivan Maillard
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Martin Carroll
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles Nelson
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Kai Tan
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Center for Single Cell Biology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Schill RL, Visser J, Ashby ML, Li Z, Lewis KT, Morales-Hernandez A, Hoose KS, Maung JN, Uranga RM, Hariri H, Hermsmeyer IDK, Mori H, MacDougald OA. Deficiency of glucocorticoid receptor in bone marrow adipocytes has mild effects on bone and hematopoiesis but does not influence expansion of marrow adiposity with caloric restriction. Front Endocrinol (Lausanne) 2024; 15:1397081. [PMID: 38887268 PMCID: PMC11180776 DOI: 10.3389/fendo.2024.1397081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
Introduction Unlike white adipose tissue depots, bone marrow adipose tissue (BMAT) expands during caloric restriction (CR). Although mechanisms for BMAT expansion remain unclear, prior research suggested an intermediary role for increased circulating glucocorticoids. Methods In this study, we utilized a recently described mouse model (BMAd-Cre) to exclusively target bone marrow adipocytes (BMAds) for elimination of the glucocorticoid receptor (GR) (i.e. Nr3c1) whilst maintaining GR expression in other adipose depots. Results Mice lacking GR in BMAds (BMAd-Nr3c1 -/-) and control mice (BMAd-Nr3c1 +/+) were fed ad libitum or placed on a 30% CR diet for six weeks. On a normal chow diet, tibiae of female BMAd-Nr3c1-/- mice had slightly elevated proximal trabecular metaphyseal bone volume fraction and thickness. Both control and BMAd-Nr3c1-/- mice had increased circulating glucocorticoids and elevated numbers of BMAds in the proximal tibia following CR. However, no significant differences in trabecular and cortical bone were observed, and quantification with osmium tetroxide and μCT revealed no difference in BMAT accumulation between control or BMAd-Nr3c1 -/- mice. Differences in BMAd size were not observed between BMAd-Nr3c1-/- and control mice. Interestingly, BMAd-Nr3c1-/- mice had decreased circulating white blood cell counts 4 h into the light cycle. Discussion In conclusion, our data suggest that eliminating GR from BMAd has minor effects on bone and hematopoiesis, and does not impair BMAT accumulation during CR.
Collapse
Affiliation(s)
- Rebecca L. Schill
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Jack Visser
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Mariah L. Ashby
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Ziru Li
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Kenneth T. Lewis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Antonio Morales-Hernandez
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Keegan S. Hoose
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Jessica N. Maung
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Romina M. Uranga
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Hadla Hariri
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Isabel D. K. Hermsmeyer
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Hiroyuki Mori
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Ormond A. MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
13
|
Zhou XC, Ni GX. O-linked β-N-acetylglucosaminylation may be a key regulatory factor in promoting osteogenic differentiation of bone marrow mesenchymal stromal cells. World J Stem Cells 2024; 16:228-231. [PMID: 38577231 PMCID: PMC10989286 DOI: 10.4252/wjsc.v16.i3.228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/02/2024] [Accepted: 02/29/2024] [Indexed: 03/25/2024] Open
Abstract
Cumulative evidence suggests that O-linked β-N-acetylglucosaminylation (O-GlcNAcylation) plays an important regulatory role in pathophysiological processes. Although the regulatory mechanisms of O-GlcNAcylation in tumors have been gradually elucidated, the potential mechanisms of O-GlcNAcylation in bone metabolism, particularly, in the osteogenic differentiation of bone marrow mesenchymal stromal cells (BMSCs) remains unexplored. In this study, the literature related to O-GlcNAcylation and BMSC osteogenic differentiation was reviewed, assuming that it could trigger more scholars to focus on research related to O-GlcNAcylation and bone metabolism and provide insights into the development of novel therapeutic targets for bone metabolism disorders such as osteoporosis.
Collapse
Affiliation(s)
- Xu-Chang Zhou
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Guo-Xin Ni
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, Fujian Province, China.
| |
Collapse
|
14
|
Karima G, Kim HD. Unlocking the regenerative key: Targeting stem cell factors for bone renewal. J Tissue Eng 2024; 15:20417314241287491. [PMID: 39479284 PMCID: PMC11523181 DOI: 10.1177/20417314241287491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/12/2024] [Indexed: 11/02/2024] Open
Abstract
Stem cell factors (SCFs) are pivotal factors existing in both soluble and membrane-bound forms, expressed by endothelial cells (ECs) and fibroblasts throughout the body. These factors enhance cell growth, viability, and migration in multipotent cell lineages. The preferential expression of SCF by arteriolar ECs indicates that arterioles create a unique microenvironment tailored to hematopoietic stem cells (HSCs). Insufficiency of SCF within bone marrow (BM)-derived adipose tissue results in decreased their overall cellularity, affecting HSCs and their immediate progenitors critical for generating diverse blood cells and maintaining the hematopoietic microenvironment. SCF deficiency disrupts BM function, impacting the production and differentiation of HSCs. Additionally, deleting SCF from adipocytes reduces lipogenesis, highlighting the crucial role of SCF/c-kit signaling in controlling lipid accumulation. This review elucidates the sources, roles, mechanisms, and molecular strategies of SCF in bone renewal, offering a comprehensive overview of recent advancements, challenges, and future directions for leveraging SCF as a key agent in regenerative medicine.
Collapse
Affiliation(s)
- Gul Karima
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| | - Hwan D. Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, Republic of Korea
- Department of IT Convergence (Brain Korea Plus 21), Korea National University of Transportation, Chungju, Republic of Korea
- Department of Biomedical Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| |
Collapse
|
15
|
Zhang S, Paccalet A, Rohde D, Cremer S, Hulsmans M, Lee IH, Mentkowski K, Grune J, Schloss MJ, Honold L, Iwamoto Y, Zheng Y, Bredella MA, Buckless C, Ghoshhajra B, Thondapu V, van der Laan AM, Piek JJ, Niessen HWM, Pallante F, Carnevale R, Perrotta S, Carnevale D, Iborra-Egea O, Muñoz-Guijosa C, Galvez-Monton C, Bayes-Genis A, Vidoudez C, Trauger SA, Scadden D, Swirski FK, Moskowitz MA, Naxerova K, Nahrendorf M. Bone marrow adipocytes fuel emergency hematopoiesis after myocardial infarction. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1277-1290. [PMID: 38344689 PMCID: PMC10857823 DOI: 10.1038/s44161-023-00388-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 02/15/2024]
Abstract
After myocardial infarction (MI), emergency hematopoiesis produces inflammatory myeloid cells that accelerate atherosclerosis and promote heart failure. Since the balance between glycolysis and mitochondrial metabolism regulates hematopoietic stem cell homeostasis, metabolic cues may influence emergency myelopoiesis. Here, we show in humans and female mice that hematopoietic progenitor cells increase fatty acid metabolism after MI. Blockade of fatty acid oxidation by deleting carnitine palmitoyltransferase (Cpt1A) in hematopoietic cells of Vav1Cre/+Cpt1Afl/fl mice limited hematopoietic progenitor proliferation and myeloid cell expansion after MI. We also observed reduced bone marrow adiposity in humans, pigs and mice following MI. Inhibiting lipolysis in adipocytes using AdipoqCreERT2Atglfl/fl mice or local depletion of bone marrow adipocytes in AdipoqCreERT2iDTR mice also curbed emergency hematopoiesis. Furthermore, systemic and regional sympathectomy prevented bone marrow adipocyte shrinkage after MI. These data establish a critical role for fatty acid metabolism in post-MI emergency hematopoiesis.
Collapse
Affiliation(s)
- Shuang Zhang
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Alexandre Paccalet
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - David Rohde
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sebastian Cremer
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Maarten Hulsmans
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - I-Hsiu Lee
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kyle Mentkowski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jana Grune
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Maximilian J Schloss
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lisa Honold
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yi Zheng
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Miriam A Bredella
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Colleen Buckless
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian Ghoshhajra
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Vikas Thondapu
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Anja M van der Laan
- Department of Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan J Piek
- Department of Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Hans W M Niessen
- Department of Pathology and Cardiac Surgery, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VU Medical Center, Amsterdam, The Netherlands
| | - Fabio Pallante
- Department of AngioCardioNeurology and Translational Medicine, I.R.C.C.S. INM Neuromed, Pozzilli, Italy
| | - Raimondo Carnevale
- Department of AngioCardioNeurology and Translational Medicine, I.R.C.C.S. INM Neuromed, Pozzilli, Italy
| | - Sara Perrotta
- Department of AngioCardioNeurology and Translational Medicine, I.R.C.C.S. INM Neuromed, Pozzilli, Italy
| | - Daniela Carnevale
- Department of AngioCardioNeurology and Translational Medicine, I.R.C.C.S. INM Neuromed, Pozzilli, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | - Charles Vidoudez
- Harvard Center for Mass Spectrometry, Harvard University, Cambridge, MA, USA
| | - Sunia A Trauger
- Harvard Center for Mass Spectrometry, Harvard University, Cambridge, MA, USA
| | - David Scadden
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Filip K Swirski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael A Moskowitz
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kamila Naxerova
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
16
|
Li Z, Rosen CJ. The Multifaceted Roles of Bone Marrow Adipocytes in Bone and Hematopoietic Homeostasis. J Clin Endocrinol Metab 2023; 108:e1465-e1472. [PMID: 37315208 PMCID: PMC12102716 DOI: 10.1210/clinem/dgad355] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/16/2023]
Abstract
Bone marrow adipose tissue (BMAT) makes up a significant portion of the marrow space, ranging from 50% to 70%, in healthy adults. It expands with aging, obesity, anorexia nervosa, and irradiation, which are conditions associated with skeletal complications or hematopoietic disorders. Therefore, BMAT has been viewed as a negative component of the bone marrow niche for decades, although the mechanisms and causative relationships have not been well-addressed. Of note, recent studies have revealed that BMAT is a multifaceted tissue that can serve as an energy reservoir to fuel osteoblasts and hematopoietic cells under stressful situations, and also acts as an endocrine/paracrine organ to suppress bone formation and support hematopoiesis at steady-state conditions. In this review, we summarize the uniqueness of BMAT, the complex findings of previous studies, and update our understanding of the physiological roles of BMAT in bone and hematopoietic metabolism based on a newly established bone marrow adipocyte-specific mouse model.
Collapse
Affiliation(s)
- Ziru Li
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA
| | - Clifford J Rosen
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA
| |
Collapse
|
17
|
Todosenko N, Khaziakhmatova O, Malashchenko V, Yurova K, Bograya M, Beletskaya M, Vulf M, Mikhailova L, Minchenko A, Soroko I, Khlusov I, Litvinova L. Adipocyte- and Monocyte-Mediated Vicious Circle of Inflammation and Obesity (Review of Cellular and Molecular Mechanisms). Int J Mol Sci 2023; 24:12259. [PMID: 37569635 PMCID: PMC10418857 DOI: 10.3390/ijms241512259] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Monocytes play a key role in the development of metabolic syndrome, and especially obesity. Given the complex features of their development from progenitor cells, whose regulation is mediated by their interactions with bone marrow adipocytes, the importance of a detailed study of the heterogeneous composition of monocytes at the molecular and systemic levels becomes clear. Research argues for monocytes as indicators of changes in the body's metabolism and the possibility of developing therapeutic strategies to combat obesity and components of metabolic syndrome based on manipulations of the monocyte compound of the immune response. An in-depth study of the heterogeneity of bone-marrow-derived monocytes and adipocytes could provide answers to many questions about the pathogenesis of obesity and reveal their therapeutic potential.
Collapse
Affiliation(s)
- Natalia Todosenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (L.M.); (A.M.); (I.S.); (I.K.)
| | - Olga Khaziakhmatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (L.M.); (A.M.); (I.S.); (I.K.)
| | - Vladimir Malashchenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (L.M.); (A.M.); (I.S.); (I.K.)
| | - Kristina Yurova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (L.M.); (A.M.); (I.S.); (I.K.)
| | - Maria Bograya
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (L.M.); (A.M.); (I.S.); (I.K.)
| | - Maria Beletskaya
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (L.M.); (A.M.); (I.S.); (I.K.)
| | - Maria Vulf
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (L.M.); (A.M.); (I.S.); (I.K.)
| | - Larisa Mikhailova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (L.M.); (A.M.); (I.S.); (I.K.)
| | - Anastasia Minchenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (L.M.); (A.M.); (I.S.); (I.K.)
| | - Irina Soroko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (L.M.); (A.M.); (I.S.); (I.K.)
| | - Igor Khlusov
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (L.M.); (A.M.); (I.S.); (I.K.)
- Laboratory of Cellular and Microfluidic Technologies, Siberian State Medical University, 2, Moskovskii Trakt, 634050 Tomsk, Russia
| | - Larisa Litvinova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (L.M.); (A.M.); (I.S.); (I.K.)
- Laboratory of Cellular and Microfluidic Technologies, Siberian State Medical University, 2, Moskovskii Trakt, 634050 Tomsk, Russia
| |
Collapse
|
18
|
Moein S, Ahmadbeigi N, Adibi R, Kamali S, Moradzadeh K, Nematollahi P, Nardi NB, Gheisari Y. Regenerative potential of multinucleated cells: bone marrow adiponectin-positive multinucleated cells take the lead. Stem Cell Res Ther 2023; 14:173. [PMID: 37403181 DOI: 10.1186/s13287-023-03400-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: 08/08/2022] [Accepted: 06/13/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Polyploid cells can be found in a wide evolutionary spectrum of organisms. These cells are assumed to be involved in tissue regeneration and resistance to stressors. Although the appearance of large multinucleated cells (LMCs) in long-term culture of bone marrow (BM) mesenchymal cells has been reported, the presence and characteristics of such cells in native BM and their putative role in BM reconstitution following injury have not been fully investigated. METHODS BM-derived LMCs were explored by time-lapse microscopy from the first hours post-isolation to assess their colony formation and plasticity. In addition, sub-lethally irradiated mice were killed every other day for four weeks to investigate the histopathological processes during BM regeneration. Moreover, LMCs from GFP transgenic mice were transplanted to BM-ablated recipients to evaluate their contribution to tissue reconstruction. RESULTS BM-isolated LMCs produced mononucleated cells with characteristics of mesenchymal stromal cells. Time-series inspections of BM sections following irradiation revealed that LMCs are highly resistant to injury and originate mononucleated cells which reconstitute the tissue. The regeneration process was synchronized with a transient augmentation of adipocytes suggesting their contribution to tissue repair. Additionally, LMCs were found to be adiponectin positive linking the observations on multinucleation and adipogenesis to BM regeneration. Notably, transplantation of LMCs to myeloablated recipients could reconstitute both the hematopoietic system and BM stroma. CONCLUSIONS A population of resistant multinucleated cells reside in the BM that serves as the common origin of stromal and hematopoietic lineages with a key role in tissue regeneration. Furthermore, this study underscores the contribution of adipocytes in BM reconstruction.
Collapse
Affiliation(s)
- Shiva Moein
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Naser Ahmadbeigi
- Gene Therapy Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Rezvan Adibi
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sara Kamali
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran
| | - Kobra Moradzadeh
- Gene Therapy Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Pardis Nematollahi
- Department of Pathology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nance Beyer Nardi
- Institute of Cardiology of Rio Grande do Sul, Av Princesa Isabel 370, Porto Alegre, RS, 90620-001, Brazil
| | - Yousof Gheisari
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran.
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
19
|
Jacobi H, Vieri M, Bütow M, Namasu CY, Flüter L, Costa IG, Maié T, Lindemann-Docter K, Chatain N, Beier F, Huber M, Wagner W, Crysandt M, Brümmendorf TH, Schemionek M. Myelofibrosis at diagnosis is associated with the failure of treatment-free remission in CML patients. Front Pharmacol 2023; 14:1212392. [PMID: 37469867 PMCID: PMC10352620 DOI: 10.3389/fphar.2023.1212392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023] Open
Abstract
The management of patients with chronic myeloid leukemia (CML) has been revolutionized by the introduction of tyrosine kinase inhibitors (TKIs), which induce deep molecular responses so that treatment can eventually be discontinued, leading to treatment-free remission (TFR) in a subset of patients. Unfortunately, leukemic stem cells (LSCs) often persist and a fraction of these can again expand in about half of patients that attempt TKI discontinuation. In this study, we show that presence of myelofibrosis (MF) at the time of diagnosis is a factor associating with TFR failure. Fibrotic transformation is governed by the action of several cytokines, and interestingly, some of them have also been described to support LSC persistence. At the cellular level, these could be produced by both malignant cells and by components of the bone marrow (BM) niche, including megakaryocytes (MKs) and mesenchymal stromal cells (MSCs). In our cohort of 57 patients, around 40% presented with MF at diagnosis and the number of blasts in the peripheral blood and BM was significantly elevated in patients with higher grade of MF. Employing a CML transgenic mouse model, we could observe higher levels of alpha-smooth muscle actin (α-SMA) in the BM when compared to control mice. Short-term treatment with the TKI nilotinib, efficiently reduced spleen weight and BCR::ABL1 mRNA levels, while α-SMA expression was only partially reduced. Interestingly, the number of MKs was increased in the spleen of CML mice and elevated in both BM and spleen upon nilotinib treatment. Analysis of human CML-vs healthy donor (HD)-derived MSCs showed an altered expression of gene signatures reflecting fibrosis as well as hematopoietic support, thus suggesting MSCs as a potential player in these two processes. Finally, in our cohort, 12 patients qualified for TKI discontinuation, and here we observed that all patients who failed TFR had BM fibrosis at diagnosis, whereas this was only the case in 25% of patients with achieved TFR, further supporting the link between fibrosis and LSC persistence.
Collapse
Affiliation(s)
- Henrike Jacobi
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Margherita Vieri
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Marlena Bütow
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Carolina Y. Namasu
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Laura Flüter
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Ivan G. Costa
- Institute for Computational Genomics, RWTH Aachen University, Aachen, Germany
| | - Tiago Maié
- Institute for Computational Genomics, RWTH Aachen University, Aachen, Germany
| | | | - Nicolas Chatain
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Fabian Beier
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, RWTH Aachen University, Aachen, Germany
| | - Wolfgang Wagner
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Institute for Stem Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
| | - Martina Crysandt
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Tim H. Brümmendorf
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Mirle Schemionek
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| |
Collapse
|
20
|
Austin MJ, Kalampalika F, Cawthorn WP, Patel B. Turning the spotlight on bone marrow adipocytes in haematological malignancy and non-malignant conditions. Br J Haematol 2023; 201:605-619. [PMID: 37067783 PMCID: PMC10952811 DOI: 10.1111/bjh.18748] [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: 12/20/2022] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 04/18/2023]
Abstract
Whilst bone marrow adipocytes (BMAd) have long been appreciated by clinical haemato-pathologists, it is only relatively recently, in the face of emerging data, that the adipocytic niche has come under the watchful eye of biologists. There is now mounting evidence to suggest that BMAds are not just a simple structural entity of bone marrow microenvironments but a bona fide driver of physio- and pathophysiological processes relevant to multiple aspects of health and disease. Whilst the truly multifaceted nature of BMAds has only just begun to emerge, paradigms have shifted already for normal, malignant and non-malignant haemopoiesis incorporating a view of adipocyte regulation. Major efforts are ongoing, to delineate the routes by which BMAds participate in health and disease with a final aim of achieving clinical tractability. This review summarises the emerging role of BMAds across the spectrum of normal and pathological haematological conditions with a particular focus on its impact on cancer therapy.
Collapse
Affiliation(s)
- Michael J. Austin
- Barts Cancer Institute, Centre for Haemato‐OncologyQueen Mary University of LondonLondonUK
| | - Foteini Kalampalika
- Barts Cancer Institute, Centre for Haemato‐OncologyQueen Mary University of LondonLondonUK
| | - William P. Cawthorn
- BHF/University Centre for Cardiovascular Science, Edinburgh BioquarterUniversity of EdinburghEdinburghUK
| | - Bela Patel
- Barts Cancer Institute, Centre for Haemato‐OncologyQueen Mary University of LondonLondonUK
| |
Collapse
|
21
|
Bone Marrow Adipose Tissue: Regulation of Osteoblastic Niche, Hematopoiesis and Hematological Malignancies. Stem Cell Rev Rep 2023:10.1007/s12015-023-10531-3. [PMID: 36930385 DOI: 10.1007/s12015-023-10531-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2023] [Indexed: 03/18/2023]
Abstract
Bone marrow adipose tissue (BMAT) creates a specific microniche within multifunctional bone marrow (BM) ecosystem which imposes changes in surrounding cells and at systemic level. Moreover, BMAT contributes to spatial and temporal separation and metabolic compartmentalization of BM, thus regulating BM homeostasis and diseases. Recent findings have identified novel progenitor subsets of bone marrow adipocytes (BMAd)s recruited during the BM adipogenesis within different skeletal and hematopoietic stem cell niches. Potential of certain mesenchymal BM cells to differentiate into both osteogenic and adipogenic lineages, contributes to the complex interplay of BMAT with endosteal (osteoblastic) niche compartments as an important cellular player in bone tissue homeostasis. Targeting and ablation of BMAT cells at certain states might be an optional and promising strategy for improvement of bone health. Additionally, recent findings demonstrated spatial distribution of BMAds related to hematopoietic cells and pointed out important functional roles in the vital processes such as long-term hematopoiesis. BM adipogenesis appears to be an emergency phenomenon that follows the production of hematopoietic stem and progenitor cell niche factors, thus regulating physiological, stressed, and malignant hematopoiesis. Lipolytic and secretory activity of BMAds can influence survival and proliferation of hematopoietic cells at different maturation stages. Due to their different lipid status, constitutive and regulated BMAds are important determinants of normal and malignant hematopoietic cells. Further elucidation of cellular and molecular players involved in BMAT expansion and crosstalk with malignant cells is of paramount importance for conceiving the new therapies for improvement of BM health.
Collapse
|
22
|
Zhang Z, Huang Z, Awad M, Elsalanty M, Cray J, Ball LE, Maynard JC, Burlingame AL, Zeng H, Mansky KC, Ruan HB. O-GlcNAc glycosylation orchestrates fate decision and niche function of bone marrow stromal progenitors. eLife 2023; 12:e85464. [PMID: 36861967 PMCID: PMC10032655 DOI: 10.7554/elife.85464] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/01/2023] [Indexed: 03/03/2023] Open
Abstract
In mammals, interactions between the bone marrow (BM) stroma and hematopoietic progenitors contribute to bone-BM homeostasis. Perinatal bone growth and ossification provide a microenvironment for the transition to definitive hematopoiesis; however, mechanisms and interactions orchestrating the development of skeletal and hematopoietic systems remain largely unknown. Here, we establish intracellular O-linked β-N-acetylglucosamine (O-GlcNAc) modification as a posttranslational switch that dictates the differentiation fate and niche function of early BM stromal cells (BMSCs). By modifying and activating RUNX2, O-GlcNAcylation promotes osteogenic differentiation of BMSCs and stromal IL-7 expression to support lymphopoiesis. In contrast, C/EBPβ-dependent marrow adipogenesis and expression of myelopoietic stem cell factor (SCF) is inhibited by O-GlcNAcylation. Ablating O-GlcNAc transferase (OGT) in BMSCs leads to impaired bone formation, increased marrow adiposity, as well as defective B-cell lymphopoiesis and myeloid overproduction in mice. Thus, the balance of osteogenic and adipogenic differentiation of BMSCs is determined by reciprocal O-GlcNAc regulation of transcription factors, which simultaneously shapes the hematopoietic niche.
Collapse
Affiliation(s)
- Zengdi Zhang
- Department of Integrative Biology and Physiology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Zan Huang
- Department of Integrative Biology and Physiology, University of Minnesota Medical SchoolMinneapolisUnited States
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural UniversityNanjingChina
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural UniversityNanjingChina
| | - Mohamed Awad
- Department of Medical Anatomical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health SciencesPomonaUnited States
| | - Mohammed Elsalanty
- Department of Medical Anatomical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health SciencesPomonaUnited States
| | - James Cray
- Department of Biomedical Education and Anatomy, The Ohio State University College of Medicine, and Division of Biosciences, The Ohio State University College of DentistryColumbusUnited States
| | - Lauren E Ball
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South CarolinaCharlestonUnited States
| | - Jason C Maynard
- Department of Pharmaceutical Chemistry, University of California, San FranciscoSan FranciscoUnited States
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California, San FranciscoSan FranciscoUnited States
| | - Hu Zeng
- Division of Rheumatology, Department of Internal Medicine, Mayo ClinicRochesterUnited States
- Department of Immunology, Mayo ClinicRochesterUnited States
| | - Kim C Mansky
- Department of Developmental and Surgical Sciences, School of Dentistry, University of MinnesotaMinneapolisUnited States
| | - Hai-Bin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota Medical SchoolMinneapolisUnited States
- Center for Immunology, University of Minnesota Medical SchoolMinneapolisUnited States
| |
Collapse
|
23
|
Vanhie JJ, Kim W, Ek Orloff L, Ngu M, Collao N, De Lisio M. The role of exercise-and high fat diet-induced bone marrow extracellular vesicles in stress hematopoiesis. Front Physiol 2022; 13:1054463. [PMID: 36505084 PMCID: PMC9728614 DOI: 10.3389/fphys.2022.1054463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
Abstract
Exercise and obesity regulate hematopoiesis, in part through alterations in cellular and soluble components of the bone marrow niche. Extracellular vesicles (EVs) are components of the bone marrow niche that regulate hematopoiesis; however, the role of exercise training or obesity induced EVs in regulating hematopoiesis remains unknown. To address this gap, donor EVs were isolated from control diet-fed, sedentary mice (CON-SED), control diet-fed exercise trained mice (CON-EX), high fat diet-fed, sedentary mice (HFD-SED), and high fat diet-fed, exercise trained mice (HFD-EX) and injected into recipient mice undergoing stress hematopoiesis. Hematopoietic and niche cell populations were quantified, and EV miRNA cargo was evaluated. EV content did not differ between the four groups. Mice receiving HFD-EX EVs had fewer hematopoietic stem cells (HSCs) (p < 0.01), long-term HSC (p < 0.05), multipotent progenitors (p < 0.01), common myeloid progenitors (p<0.01), common lymphoid progenitors (p < 0.01), and granulocyte-macrophage progenitors (p < 0.05), compared to mice receiving HFD-SED EVs. Similarly, mice receiving EX EVs had fewer osteoprogenitor cells compared to SED (p < 0.05) but enhanced mesenchymal stromal cell (MSC) osteogenic differentiation in vitro (p < 0.05) compared to SED EVs. HFD EVs enhanced mesenchymal stromal cell (MSC) adipogenesis in vitro (p < 0.01) compared to CON EVs. HFD-EX EVs had lower microRNA-193 and microRNA-331-5p content, microRNAs implicated in inhibiting osteogenesis and leukemic cell expansion respectively, compared to HFD-SED EVs. The results identify alterations in EV cargo as a novel mechanism by which exercise training alters stress hematopoiesis and the bone marrow niche.
Collapse
Affiliation(s)
- James J. Vanhie
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Wooseok Kim
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Lisa Ek Orloff
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Matthew Ngu
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Nicolas Collao
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Michael De Lisio
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada,*Correspondence: Michael De Lisio,
| |
Collapse
|
24
|
Li Z, Bagchi DP, Zhu J, Bowers E, Yu H, Hardij J, Mori H, Granger K, Skjaerlund J, Mandair G, Abrishami S, Singer K, Hankenson KD, Rosen CJ, MacDougald OA. Constitutive bone marrow adipocytes suppress local bone formation. JCI Insight 2022; 7:160915. [PMID: 36048537 PMCID: PMC9675472 DOI: 10.1172/jci.insight.160915] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/31/2022] [Indexed: 12/15/2022] Open
Abstract
BM adipocytes (BMAd) are a unique cell population derived from BM mesenchymal progenitors and marrow adipogenic lineage precursors. Although they have long been considered to be a space filler within bone cavities, recent studies have revealed important physiological roles in hematopoiesis and bone metabolism. To date, the approaches used to study BMAd function have been confounded by contributions by nonmarrow adipocytes or by BM stromal cells. To address this gap in the field, we have developed a BMAd-specific Cre mouse model to deplete BMAds by expression of diphtheria toxin A (DTA) or by deletion of peroxisome proliferator-activated receptor gamma (Pparg). We found that DTA-induced loss of BMAds results in decreased hematopoietic stem and progenitor cell numbers and increased bone mass in BMAd-enriched locations, including the distal tibiae and caudal vertebrae. Elevated bone mass appears to be secondary to enhanced endosteal bone formation, suggesting a local effect caused by depletion of BMAd. Augmented bone formation with BMAd depletion protects mice from bone loss induced by caloric restriction or ovariectomy, and it facilitates the bone-healing process after fracture. Finally, ablation of Pparg also reduces BMAd numbers and largely recapitulates high-bone mass phenotypes observed with DTA-induced BMAd depletion.
Collapse
Affiliation(s)
- Ziru Li
- Department of Molecular & Integrative Physiology and
| | | | - Junxiong Zhu
- Department of Orthopedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Emily Bowers
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Hui Yu
- Department of Molecular & Integrative Physiology and
| | - Julie Hardij
- Department of Molecular & Integrative Physiology and
| | - Hiroyuki Mori
- Department of Molecular & Integrative Physiology and
| | | | - Jon Skjaerlund
- Department of Orthopedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Gurjit Mandair
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Simin Abrishami
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kanakadurga Singer
- Department of Molecular & Integrative Physiology and
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kurt D. Hankenson
- Department of Orthopedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Ormond A. MacDougald
- Department of Molecular & Integrative Physiology and
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
25
|
Vauclard A, Bellio M, Valet C, Borret M, Payrastre B, Severin S. Obesity: Effects on bone marrow homeostasis and platelet activation. Thromb Res 2022. [DOI: 10.1016/j.thromres.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
26
|
Bone Marrow Fat Distribution in Patients With β-Thalassemia: A Study Using Chemical Shift-Based Water-Fat MRI. Acad Radiol 2022; 29:e39-e48. [PMID: 33992535 DOI: 10.1016/j.acra.2021.03.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 11/01/2022]
Abstract
RATIONALE AND OBJECTIVES Molecular studies have shown the changes in bone marrow fat in relation to altered hematopoiesis. This study aims to investigate the changes in the bone marrow fat in patients affected by β-thalassemia by using chemical shift-encoded (CSE)-MRI. MATERIALS AND METHODS Twenty-three subjects, comprising of six healthy (17-31 years old) and 17 β-thalassemia subjects (19-39 years old), were scanned using a multiecho fast low angle shot sequence (0.94 × 0.94 × 3.00 mm3) and a stimulated echo acquisition mode sequence using 3T MRI. Bone marrow proton density fat fraction (PDFF) was quantified in the left femur of each subject. Regression and Bland-Altman analysis were used to analyze agreement between CSE-MRI and 1H-MRS. PDFF distribution was analyzed using Hartigan's dip test and the computed Wasserstein distances. Jonckheere-Terpstra trend analysis was performed to evaluate the effect of disease severity on PDFF distribution. RESULTS An excellent agreement was found between PDFF measured using CSE-MRI with 1H-MRS (R2 = 0.91; bias =-1.41%). Healthy subjects showed left-skewed or bimodal PDFF distribution while β-thalassemia subjects showed bimodal, normal or right-skewed distribution. Jonckheere-Terpstra test shows that PDFF distribution was increasingly different from the norm as disease severity increased (TJT = 166.0, z = 3.806, p < 0.05). Increase in variability of PDFF distribution within each subject group was also seen with increasing disease severity (TJT = 169.0, z = 3.971, p < 0.05). CONCLUSION CSE-MRI is a promising tool to demonstrate spatial changes and variability in marrow fat distribution, resulting from ineffective erythropoiesis.
Collapse
|
27
|
Little-Letsinger SE, Rubin J, Diekman B, Rubin CT, McGrath C, Pagnotti GM, Klett EL, Styner M. Exercise to Mend Aged-tissue Crosstalk in Bone Targeting Osteoporosis & Osteoarthritis. Semin Cell Dev Biol 2022; 123:22-35. [PMID: 34489173 PMCID: PMC8840966 DOI: 10.1016/j.semcdb.2021.08.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 12/16/2022]
Abstract
Aging induces alterations in bone structure and strength through a multitude of processes, exacerbating common aging- related diseases like osteoporosis and osteoarthritis. Cellular hallmarks of aging are examined, as related to bone and the marrow microenvironment, and ways in which these might contribute to a variety of age-related perturbations in osteoblasts, osteocytes, marrow adipocytes, chondrocytes, osteoclasts, and their respective progenitors. Cellular senescence, stem cell exhaustion, mitochondrial dysfunction, epigenetic and intracellular communication changes are central pathways and recognized as associated and potentially causal in aging. We focus on these in musculoskeletal system and highlight knowledge gaps in the literature regarding cellular and tissue crosstalk in bone, cartilage, and the bone marrow niche. While senolytics have been utilized to target aging pathways, here we propose non-pharmacologic, exercise-based interventions as prospective "senolytics" against aging effects on the skeleton. Increased bone mass and delayed onset or progression of osteoporosis and osteoarthritis are some of the recognized benefits of regular exercise across the lifespan. Further investigation is needed to delineate how cellular indicators of aging manifest in bone and the marrow niche and how altered cellular and tissue crosstalk impact disease progression, as well as consideration of exercise as a therapeutic modality, as a means to enhance discovery of bone-targeted therapies.
Collapse
Affiliation(s)
- SE Little-Letsinger
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill
| | - J Rubin
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill,North Carolina Diabetes Research Center (NCDRC), University of North Carolina at Chapel Hill,Department of Medicine, Thurston Arthritis Research Center (TARC), University of North Carolina at Chapel Hill
| | - B Diekman
- Department of Medicine, Thurston Arthritis Research Center (TARC), University of North Carolina at Chapel Hill,Joint Departments of Biomedical Engineering NC State & University of North Carolina at Chapel Hill
| | - CT Rubin
- Department of Biomedical Engineering, State University of New York at Stony Brook
| | - C McGrath
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill
| | - GM Pagnotti
- Dept of Endocrine, Neoplasia, and Hormonal Disorders, University Texas MD Anderson Cancer Center, Houston
| | - EL Klett
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill,Department of Nutrition, School of Public Health, University of North Carolina at Chapel Hill
| | - M Styner
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill,North Carolina Diabetes Research Center (NCDRC), University of North Carolina at Chapel Hill,Department of Medicine, Thurston Arthritis Research Center (TARC), University of North Carolina at Chapel Hill
| |
Collapse
|
28
|
Zingariello M, Verachi P, Gobbo F, Martelli F, Falchi M, Mazzarini M, Valeri M, Sarli G, Marinaccio C, Melo-Cardenas J, Crispino JD, Migliaccio AR. Resident Self-Tissue of Proinflammatory Cytokines Rather than Their Systemic Levels Correlates with Development of Myelofibrosis in Gata1low Mice. Biomolecules 2022; 12:biom12020234. [PMID: 35204735 PMCID: PMC8961549 DOI: 10.3390/biom12020234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/27/2022] [Accepted: 01/27/2022] [Indexed: 02/05/2023] Open
Abstract
Serum levels of inflammatory cytokines are currently investigated as prognosis markers in myelofibrosis, the most severe Philadelphia-negative myeloproliferative neoplasm. We tested this hypothesis in the Gata1low model of myelofibrosis. Gata1low mice, and age-matched wild-type littermates, were analyzed before and after disease onset. We assessed cytokine serum levels by Luminex-bead-assay and ELISA, frequency and cytokine content of stromal cells by flow cytometry, and immunohistochemistry and bone marrow (BM) localization of GFP-tagged hematopoietic stem cells (HSC) by confocal microscopy. Differences in serum levels of 32 inflammatory-cytokines between prefibrotic and fibrotic Gata1low mice and their wild-type littermates were modest. However, BM from fibrotic Gata1low mice contained higher levels of lipocalin-2, CXCL1, and TGF-β1 than wild-type BM. Although frequencies of endothelial cells, mesenchymal cells, osteoblasts, and megakaryocytes were higher than normal in Gata1low BM, the cells which expressed these cytokines the most were malignant megakaryocytes. This increased bioavailability of proinflammatory cytokines was associated with altered HSC localization: Gata1low HSC were localized in the femur diaphysis in areas surrounded by microvessels, neo-bones, and megakaryocytes, while wild-type HSC were localized in the femur epiphysis around adipocytes. In conclusion, bioavailability of inflammatory cytokines in BM, rather than blood levels, possibly by reshaping the HSC niche, correlates with myelofibrosis in Gata1low mice.
Collapse
Affiliation(s)
| | - Paola Verachi
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
| | - Francesca Gobbo
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Mario Falchi
- National Center HIV/AIDS Research, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Maria Mazzarini
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
| | - Mauro Valeri
- Center for Animal Experimentation and Well-Being, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy;
| | | | - Johanna Melo-Cardenas
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.M.-C.); (J.D.C.)
| | - John D. Crispino
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.M.-C.); (J.D.C.)
| | - Anna Rita Migliaccio
- Altius Institute for Biomedical Sciences, Seattle, WA 98121, USA
- Center for Integrated Biomedical Research, Campus Bio-Medico, 00128 Rome, Italy
- Correspondence:
| |
Collapse
|
29
|
Turner RT, Nesser KL, Philbrick KA, Wong CP, Olson DA, Branscum AJ, Iwaniec UT. Leptin and environmental temperature as determinants of bone marrow adiposity in female mice. Front Endocrinol (Lausanne) 2022; 13:959743. [PMID: 36277726 PMCID: PMC9582271 DOI: 10.3389/fendo.2022.959743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/15/2022] [Indexed: 11/21/2022] Open
Abstract
Bone marrow adipose tissue (BMAT) levels are higher in distal femur metaphysis of female mice housed at thermoneutral (32°C) than in mice housed at 22°C, as are abdominal white adipose tissue (WAT) mass, and serum leptin levels. We performed two experiments to explore the role of increased leptin in temperature-enhanced accrual of BMAT. First, we supplemented 6-week-old female C57BL/6J (B6) mice with leptin for 2 weeks at 10 µg/d using a subcutaneously implanted osmotic pump. Controls consisted of ad libitum (ad lib) fed mice and mice pair fed to match food intake of leptin-supplemented mice. The mice were maintained at 32°C for the duration of treatment. At necropsy, serum leptin in leptin-supplemented mice did not differ from ad lib mice, suggesting suppression of endogenous leptin production. In support, Ucp1 expression in BAT, percent body fat, and abdominal WAT mass were lower in leptin-supplemented mice. Leptin-supplemented mice also had lower BMAT and higher bone formation in distal femur metaphysis compared to the ad lib group, changes not replicated by pair-feeding. In the second experiment, BMAT response was evaluated in 6-week-old female B6 wild type (WT), leptin-deficient ob/ob and leptin-treated (0.3 μg/d) ob/ob mice housed at 32°C for the 2-week duration of the treatment. Compared to mice sacrificed at baseline (22°C), BMAT increased in ob/ob mice as well as WT mice, indicating a leptin independent response to increased temperature. However, infusion of ob/ob mice with leptin, at a dose rate having negligible effects on either energy metabolism or serum leptin levels, attenuated the increase in BMAT. In summary, increased housing temperature and increased leptin have independent but opposing effects on BMAT in mice.
Collapse
Affiliation(s)
- Russell T. Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, United States
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR, United States
| | - Kira L. Nesser
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, United States
| | - Kenneth A. Philbrick
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, United States
| | - Carmen P. Wong
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, United States
| | - Dawn A. Olson
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, United States
| | - Adam J. Branscum
- Biostatistics Program, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, United States
| | - Urszula T. Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, United States
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR, United States
- *Correspondence: Urszula T. Iwaniec,
| |
Collapse
|
30
|
Tratwal J, Falgayrac G, During A, Bertheaume N, Bataclan C, Tavakol DN, Campos V, Duponchel L, Daley GQ, Penel G, Chauveau C, Naveiras O. Raman microspectroscopy reveals unsaturation heterogeneity at the lipid droplet level and validates an in vitro model of bone marrow adipocyte subtypes. Front Endocrinol (Lausanne) 2022; 13:1001210. [PMID: 36506047 PMCID: PMC9727239 DOI: 10.3389/fendo.2022.1001210] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/04/2022] [Indexed: 11/24/2022] Open
Abstract
Bone marrow adipocytes (BMAds) constitute the most abundant stromal component of adult human bone marrow. Two subtypes of BMAds have been described, the more labile regulated adipocytes (rBMAds) and the more stable constitutive adipocytes (cBMAds), which develop earlier in life and are more resilient to environmental and metabolic disruptions. In vivo, rBMAds are enriched in saturated fatty acids, contain smaller lipid droplets (LDs) and more readily provide hematopoietic support than their cBMAd counterparts. Mouse models have been used for BMAds research, but isolation of primary BMAds presents many challenges, and thus in vitro models remain the current standard to study nuances of adipocyte differentiation. No in vitro model has yet been described for the study of rBMAds/cBMAds. Here, we present an in vitro model of BM adipogenesis with differential rBMAd and cBMAd-like characteristics. We used OP9 BM stromal cells derived from a (C57BL/6xC3H)F2-op/op mouse, which have been extensively characterized as feeder layer for hematopoiesis research. We observed similar canonical adipogenesis transcriptional signatures for spontaneously-differentiated (sOP9) and induced (iOP9) cultures, while fatty acid composition and desaturase expression of Scd1 and Fads2 differed at the population level. To resolve differences at the single adipocyte level we tested Raman microspectroscopy and show it constitutes a high-resolution method for studying adipogenesis in vitro in a label-free manner, with resolution to individual LDs. We found sOP9 adipocytes have lower unsaturation ratios, smaller LDs and higher hematopoietic support than iOP9 adipocytes, thus functionally resembling rBMAds, while iOP9 more closely resembled cBMAds. Validation in human primary samples confirmed a higher unsaturation ratio for lipids extracted from stable cBMAd-rich sites (femoral head upon hip-replacement surgery) versus labile rBMAds (iliac crest after chemotherapy). As a result, the 16:1/16:0 fatty acid unsaturation ratio, which was already shown to discriminate BMAd subtypes in rabbit and rat marrow, was validated to discriminate cBMAds from rBMAd in both the OP9 model in vitro system and in human samples. We expect our model will be useful for cBMAd and rBMAd studies, particularly where isolation of primary BMAds is a limiting step.
Collapse
Affiliation(s)
- Josefine Tratwal
- Laboratory of Regenerative Hematopoiesis, Ecole Polytechnique Fédérale de Lausanne (EPFL) & Department of Biomedical Sciences, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Guillaume Falgayrac
- Univ. Lille, CHU Lille, Univ. Littoral Côte d’Opale, ULR 4490 - MABLab- Marrow Adiposity Laboratory, Lille, France
| | - Alexandrine During
- Univ. Lille, CHU Lille, Univ. Littoral Côte d’Opale, ULR 4490 - MABLab- Marrow Adiposity Laboratory, Lille, France
| | - Nicolas Bertheaume
- Univ. Lille, CHU Lille, Univ. Littoral Côte d’Opale, ULR 4490 - MABLab- Marrow Adiposity Laboratory, Lille, France
| | - Charles Bataclan
- Laboratory of Regenerative Hematopoiesis, Ecole Polytechnique Fédérale de Lausanne (EPFL) & Department of Biomedical Sciences, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Daniel N. Tavakol
- Laboratory of Regenerative Hematopoiesis, Ecole Polytechnique Fédérale de Lausanne (EPFL) & Department of Biomedical Sciences, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Vasco Campos
- Laboratory of Regenerative Hematopoiesis, Ecole Polytechnique Fédérale de Lausanne (EPFL) & Department of Biomedical Sciences, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Ludovic Duponchel
- Univ. Lille, CNRS, UMR 8516 - LASIRe - Laboratoire Avancé de Spectroscopie pour les Intéractions la Réactivité et l’Environnement, Lille, France
| | - George Q. Daley
- Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, Boston, MA, United States
| | - Guillaume Penel
- Univ. Lille, CHU Lille, Univ. Littoral Côte d’Opale, ULR 4490 - MABLab- Marrow Adiposity Laboratory, Lille, France
| | - Christophe Chauveau
- Univ. Lille, CHU Lille, Univ. Littoral Côte d’Opale, ULR 4490 - MABLab- Marrow Adiposity Laboratory, Lille, France
| | - Olaia Naveiras
- Laboratory of Regenerative Hematopoiesis, Ecole Polytechnique Fédérale de Lausanne (EPFL) & Department of Biomedical Sciences, University of Lausanne (UNIL), Lausanne, Switzerland
- Service of Hematology, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of Hematology, Department of Laboratory Medicine Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- *Correspondence: Olaia Naveiras,
| |
Collapse
|
31
|
Wyst KBV, Hu HH, Peña A, Olson ML, Bailey SS, Shaibi GQ. Bone marrow adipose tissue content in Latino adolescents with prediabetes and obesity. Obesity (Silver Spring) 2021; 29:2100-2107. [PMID: 34582099 PMCID: PMC8612952 DOI: 10.1002/oby.23279] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/12/2021] [Accepted: 07/12/2021] [Indexed: 11/10/2022]
Abstract
OBJECTIVE This study aimed to examine whether total, regional, and organ fat predicts bone marrow adipose tissue (BMAT) fat content and to explore whether BMAT fat content differs by sex among Latino youth. METHODS Latino youth (n = 86; age 13.6 [1.4] years, 62% male) with obesity (BMI percentile = 98.5% [1.2%]) underwent a dual-energy x-ray absorptiometry scan to assess body composition and a magnetic resonance imaging scan to determine abdominal adiposity, liver fat, and vertebral BMAT fat content in the thoracic (average of T8-T12) and lumbar (average of L1-L5) spine. RESULTS Male youth exhibited significantly greater thoracic (male youth = 30.8% [1.4%] vs. female youth = 24.5% [2.1%], p = 0.027) and lumbar (male youth = 36.3% [1.5%] vs. female youth = 30.2% [2.2%], p = 0.038) BMAT fat content compared with female youth. Visceral adipose tissue was a significant predictor of thoracic (β = 0.434, t[86] = 3.016, p = 0.003) and lumbar (β = 0.389, t[86] = 2.677, p = 0.009) BMAT fat content, explaining 8.9% and 6.9% of the variance, respectively. Liver fat was a significant predictor of both thoracic (β = 0.487, t[86] = 4.334, p < 0.001) and lumbar (β = 0.436, t[86] = 3.793, p < 0.001) BMAT fat content, explaining 17.6% and 13.8% of the variance, respectively. CONCLUSIONS Male youth had significantly greater thoracic and lumbar BMAT fat content than female youth. Greater BMAT fat content is associated with greater liver fat and visceral adipose tissue among youth with obesity. Further investigation of the mechanistic underpinnings of BMAT may help to differentiate its metabolic and bone-related functions.
Collapse
Affiliation(s)
- Kiley B. Vander Wyst
- College of Graduate Studies, Midwestern University, Glendale, AZ
- Center for Health Promotion and Disease Prevention, Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ
| | - Houchun H. Hu
- Center for Health Promotion and Disease Prevention, Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ
- Clinical Science, Hyperfine, Inc., Guilford, CT
| | - Armando Peña
- Center for Health Promotion and Disease Prevention, Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ
| | - Micah L. Olson
- Center for Health Promotion and Disease Prevention, Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ
- Division of Pediatric Endocrinology and Diabetes, Phoenix Children’s Hospital, Phoenix, AZ
| | - Smita S. Bailey
- Center for Health Promotion and Disease Prevention, Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ
- Department of Radiology, Phoenix Children’s Hospital, Phoenix, AZ
| | - Gabriel Q. Shaibi
- Center for Health Promotion and Disease Prevention, Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ
- Division of Pediatric Endocrinology and Diabetes, Phoenix Children’s Hospital, Phoenix, AZ
- Southwest Interdisciplinary Research Center, Arizona State University, Phoenix, AZ
| |
Collapse
|
32
|
Aging, Bone Marrow and Next-Generation Sequencing (NGS): Recent Advances and Future Perspectives. Int J Mol Sci 2021; 22:ijms222212225. [PMID: 34830107 PMCID: PMC8620539 DOI: 10.3390/ijms222212225] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 12/28/2022] Open
Abstract
The aging of bone marrow (BM) remains a very imperative and alluring subject, with an ever-increasing interest among fellow scientists. A considerable amount of progress has been made in this field with the established ‘hallmarks of aging’ and continued efforts to investigate the age-related changes observed within the BM. Inflammaging is considered as a low-grade state of inflammation associated with aging, and whilst the possible mechanisms by which aging occurs are now largely understood, the processes leading to the underlying changes within aged BM remain elusive. The ability to identify these changes and detect such alterations at the genetic level are key to broadening the knowledgebase of aging BM. Next-generation sequencing (NGS) is an important molecular-level application presenting the ability to not only determine genomic base changes but provide transcriptional profiling (RNA-seq), as well as a high-throughput analysis of DNA–protein interactions (ChIP-seq). Utilising NGS to explore the genetic alterations occurring over the aging process within alterative cell types facilitates the comprehension of the molecular and cellular changes influencing the dynamics of aging BM. Thus, this review prospects the current landscape of BM aging and explores how NGS technology is currently being applied within this ever-expanding field of research.
Collapse
|
33
|
Chavakis T, Wielockx B, Hajishengallis G. Inflammatory Modulation of Hematopoiesis: Linking Trained Immunity and Clonal Hematopoiesis with Chronic Disorders. Annu Rev Physiol 2021; 84:183-207. [PMID: 34614373 DOI: 10.1146/annurev-physiol-052521-013627] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Inflammation-adapted hematopoietic stem and progenitor cells (HSPCs) have long been appreciated as key drivers of emergency myelopoiesis, thereby enabling the bone marrow to meet the elevated demand for myeloid cell generation under various stress conditions, such as systemic infection, inflammation, or myelosuppressive insults. In recent years, HSPC adaptations were associated with potential involvement in the induction of long-lived trained immunity and the emergence of clonal hematopoiesis of indeterminate potential (CHIP). Whereas trained immunity has context-dependent effects, protective in infections and tumors but potentially detrimental in chronic inflammatory diseases, CHIP increases the risk for hematological neoplastic disorders and cardiometabolic pathologies. This review focuses on the inflammatory regulation of HSPCs in the aforementioned processes and discusses how modulation of HSPC function could lead to novel therapeutic interventions. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic, Technische Universität Dresden, 01307 Dresden, Germany; ,
| | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic, Technische Universität Dresden, 01307 Dresden, Germany; ,
| | - George Hajishengallis
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6030, USA;
| |
Collapse
|
34
|
Meza-León B, Gratzinger D, Aguilar-Navarro AG, Juárez-Aguilar FG, Rebel VI, Torlakovic E, Purton LE, Dorantes-Acosta EM, Escobar-Sánchez A, Dick JE, Flores-Figueroa E. Human, mouse, and dog bone marrow show similar mesenchymal stromal cells within a distinctive microenvironment. Exp Hematol 2021; 100:41-51. [PMID: 34228982 DOI: 10.1016/j.exphem.2021.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/22/2021] [Accepted: 06/26/2021] [Indexed: 12/22/2022]
Abstract
Bone marrow stromal cells (BMSCs) are a key part of the hematopoietic niche. Mouse and human BMSCs are recognized by different markers (LepR and NGFR/CD271, respectively). However, there has not been a detailed in situ comparison of both populations within the hematopoietic microenvironment. Moreover, dog BMSCs have not been characterized in situ by any of those markers. We conducted a systematic histopathological comparison of mouse, human, and dog BMSCs within their bone marrow architecture and microenvironment. Human and dog CD271+ BMSCs had a morphology, frequency, and distribution within trabecular bone marrow similar to those of mouse LepR+ BMSCs. However, mouse bone marrow had higher cellularity and megakaryocyte content. In conclusion, highly comparable bone marrow mesenchymal stromal cell distribution among the three species establishes the validity of using mouse and dog as a surrogate experimental model of hematopoietic stem cell-BMSC interactions. However, the distinct differences in adipocyte and megakaryocyte microenvironment content of mouse bone marrow and how they might influence hematopoietic stem cell interactions as compared with humans require further study.
Collapse
Affiliation(s)
- Berenice Meza-León
- Unidad de Investigación Médica en Enfermedades Oncológicas. Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, México
| | - Dita Gratzinger
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Alicia G Aguilar-Navarro
- Unidad de Investigación Médica en Enfermedades Oncológicas. Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, México
| | - Fany G Juárez-Aguilar
- Departamento de Patología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, México
| | - Vivienne I Rebel
- Department of Cell Systems & Anatomy, The University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Emina Torlakovic
- Saskatchewan Health Authority (SHA), Saskatoon, Saskatchewan, Canada; University of Saskatchewan, Saskatchewan, Canada
| | - Louise E Purton
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia; Department of Medicine, University of Melbourne, Parkville, VIC, Australia
| | - Elisa M Dorantes-Acosta
- Biobanco de Investigación en Células Leucémicas, Hospital Infantil de México Federico Gómez, Mexico City, México
| | | | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Eugenia Flores-Figueroa
- Unidad de Investigación Médica en Enfermedades Oncológicas. Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, México; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
35
|
Zhong L, Yao L, Seale P, Qin L. Marrow adipogenic lineage precursor: A new cellular component of marrow adipose tissue. Best Pract Res Clin Endocrinol Metab 2021; 35:101518. [PMID: 33812853 PMCID: PMC8440665 DOI: 10.1016/j.beem.2021.101518] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Bone marrow mesenchymal stromal cells are a highly heterogenic cell population containing mesenchymal stem cells as well as other cell types. With the advance of single cell transcriptome analysis, several recent reports identified a prominent subpopulation of mesenchymal stromal cells that specifically express adipocyte markers but do not contain lipid droplets. We name this cell type marrow adipogenic lineage precursor, MALP, and consider it as a major cellular component of marrow adipose tissue. Here, we review the discovery of MALPs and summarize their unique features and regulatory roles in bone. We further discuss how these findings advance our understanding of bone remodeling, mesenchymal niche regulation of hematopoiesis, and marrow vasculature maintenance.
Collapse
Affiliation(s)
- Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Lutian Yao
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Patrick Seale
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
36
|
Tratwal J, Rojas-Sutterlin S, Bataclan C, Blum S, Naveiras O. Bone marrow adiposity and the hematopoietic niche: A historical perspective of reciprocity, heterogeneity, and lineage commitment. Best Pract Res Clin Endocrinol Metab 2021; 35:101564. [PMID: 34417114 DOI: 10.1016/j.beem.2021.101564] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE Here we review the current knowledge on bone marrow adipocytes (BMAds) as active contributors to the regulation of the hematopoietic niche, and as potentially pivotal players in the progression of hematological malignancies. We highlight the hierarchical and functional heterogeneity of the adipocyte lineage within the bone marrow, and how potentially different contexts dictate their interactions with hematopoietic populations. RECENT FINDINGS Growing evidence associates the adipocyte lineage with important functions in hematopoietic regulation within the BM niche. Initially proposed to serve as negative regulators of the hematopoietic microenvironment, studies have also demonstrated that BMAds positively influence the survival and maintenance of hematopoietic stem cells (HSCs). These seemingly incongruous findings may at least be partially explained by stage-specificity across the adipocytic differentiation axis and by BMAds subtypes, suggesting that the heterogeneity of these populations allows for differential context-based interactions. One such distinction relies on the location of adipocytes. Constitutive bone marrow adipose tissue (cBMAT) historically associates to the "yellow" marrow containing so-called "stable" BMAs larger in size, less responsive to stimuli, and linked to HSC quiescence. On the other hand, regulated bone marrow adipose tissue (rBMAT)-associated adipocytes, also referred to as "labile" are smaller, more responsive to hematopoietic demand and strategically situated in hematopoietically active regions of the skeleton. Here we propose a model where the effect of distinct BM stromal cell populations (BMSC) in hematopoiesis is structured along the BMSC-BMAd differentiation axis, and where the effects on HSC maintenance versus hematopoietic proliferation are segregated. In doing so, it is possible to explain how recently identified, adipocyte-primed leptin receptor-expressing, CXCL12-high adventitial reticular cells (AdipoCARs) and marrow adipose lineage precursor cells (MALPs) best support active hematopoietic cell proliferation, while adipose progenitor cells (APCs) and maturing BMAd gradually lose the capacity to support active hematopoiesis, favoring HSC quiescence. Implicated soluble mediators include MCP-1, PAI-1, NRP1, possibly DPP4 and limiting availability of CXCL12 and SCF. How remodeling occurs within the BMSC-BMAd differentiation axis is yet to be elucidated and will likely unravel a three-way regulation of the hematopoietic, bone, and adipocytic compartments orchestrated by vascular elements. The interaction of malignant hematopoietic cells with BMAds is precisely contributing to unravel specific mechanisms of remodeling. SUMMARY BMAds are important operative components of the hematopoietic microenvironment. Their heterogeneity directs their ability to exert a range of regulatory capacities in a manner dependent on their hierarchical, spatial, and biological context. This complexity highlights the importance of (i) developing experimental tools and nomenclature adapted to address stage-specificity and heterogeneity across the BMSC-BMAd differentiation axis when reporting effects in hematopoiesis, (ii) interpreting gene reporter studies within this framework, and (iii) quantifying changes in all three compartments (hematopoiesis, adiposity and bone) when addressing interdependency.
Collapse
Affiliation(s)
- Josefine Tratwal
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL) & Department of Biomedical Sciences, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Shanti Rojas-Sutterlin
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL) & Department of Biomedical Sciences, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Charles Bataclan
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL) & Department of Biomedical Sciences, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Sabine Blum
- Hematology Service, Departments of Oncology and Laboratory Medicine, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Olaia Naveiras
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL) & Department of Biomedical Sciences, University of Lausanne (UNIL), Lausanne, Switzerland; Hematology Service, Departments of Oncology and Laboratory Medicine, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland.
| |
Collapse
|
37
|
Crippa S, Santi L, Berti M, De Ponti G, Bernardo ME. Role of ex vivo Expanded Mesenchymal Stromal Cells in Determining Hematopoietic Stem Cell Transplantation Outcome. Front Cell Dev Biol 2021; 9:663316. [PMID: 34017834 PMCID: PMC8129582 DOI: 10.3389/fcell.2021.663316] [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: 02/09/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
Overall, the human organism requires the production of ∼1 trillion new blood cells per day. Such goal is achieved via hematopoiesis occurring within the bone marrow (BM) under the tight regulation of hematopoietic stem and progenitor cell (HSPC) homeostasis made by the BM microenvironment. The BM niche is defined by the close interactions of HSPCs and non-hematopoietic cells of different origin, which control the maintenance of HSPCs and orchestrate hematopoiesis in response to the body’s requirements. The activity of the BM niche is regulated by specific signaling pathways in physiological conditions and in case of stress, including the one induced by the HSPC transplantation (HSCT) procedures. HSCT is the curative option for several hematological and non-hematological diseases, despite being associated with early and late complications, mainly due to a low level of HSPC engraftment, impaired hematopoietic recovery, immune-mediated graft rejection, and graft-versus-host disease (GvHD) in case of allogenic transplant. Mesenchymal stromal cells (MSCs) are key elements of the BM niche, regulating HSPC homeostasis by direct contact and secreting several paracrine factors. In this review, we will explore the several mechanisms through which MSCs impact on the supportive activity of the BM niche and regulate HSPC homeostasis. We will further discuss how the growing understanding of such mechanisms have impacted, under a clinical point of view, on the transplantation field. In more recent years, these results have instructed the design of clinical trials to ameliorate the outcome of HSCT, especially in the allogenic setting, and when low doses of HSPCs were available for transplantation.
Collapse
Affiliation(s)
- Stefania Crippa
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ludovica Santi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Margherita Berti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giada De Ponti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Maria Ester Bernardo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy.,University Vita-Salute San Raffaele, Faculty of Medicine, Milan, Italy
| |
Collapse
|
38
|
Culliton K, Louati H, Laneuville O, Ramsay T, Trudel G. Six degrees head-down tilt bed rest caused low-grade hemolysis: a prospective randomized clinical trial. NPJ Microgravity 2021; 7:4. [PMID: 33589644 PMCID: PMC7884785 DOI: 10.1038/s41526-021-00132-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 01/13/2021] [Indexed: 01/31/2023] Open
Abstract
This study aimed to measure hemolysis before, during and after 60 days of the ground-based spaceflight analog bed rest and the effect of a nutritional intervention through a prospective randomized clinical trial. Twenty male participants were hospitalized for 88 days comprised of 14 days of ambulatory baseline, 60 days of 6° head-down tilt bed rest and 14 days of reambulation. Ten participants each received a control diet or daily polyphenol associated with omega-3, vitamin E, and selenium supplements. The primary outcome was endogenous carbon monoxide (CO) elimination measured by gas chromatography. Hemolysis was also measured with serial bilirubin, iron, transferrin saturation blood levels and serial 3-day stool collections were used to measure urobilinoid excretion using photometry. Total hemoglobin mass (tHb) was measured using CO-rebreathing. CO elimination increased after 5, 11, 30, and 57 days of bed rest: +289 ppb (95% CI 101-477 ppb; p = 0.004), +253 ppb (78-427 ppb; p = 0.007), +193 ppb (89-298 ppb; p = 0.001) and +858 ppb (670-1046 ppb; p < 0.000), respectively, compared to baseline. Bilirubin increased after 20 and 49 days of bed rest +0.8 mg/l (p = 0.013) and +1.1 mg/l (p = 0.012), respectively; and iron increased after 20 days of bed rest +10.5 µg/dl (p = 0.032). The nutritional intervention did not change CO elimination. THb was lower after 60 days of bed rest -0.9 g/kg (p = 0.001). Bed rest enhanced hemolysis as measured through all three by-products of heme oxygenase. Ongoing enhanced hemolysis over 60 days contributed to a 10% decrease in tHb mass. Modulation of red blood cell control towards increased hemolysis may be an important mechanism causing anemia in astronauts.
Collapse
Affiliation(s)
- Kathryn Culliton
- grid.412687.e0000 0000 9606 5108Department of Medicine, Division of Physical Medicine and Rehabilitation, Ottawa Hospital Research Institute, Ottawa, ON Canada
| | - Hakim Louati
- grid.412687.e0000 0000 9606 5108Department of Medicine, Division of Physical Medicine and Rehabilitation, Ottawa Hospital Research Institute, Ottawa, ON Canada
| | - Odette Laneuville
- grid.28046.380000 0001 2182 2255Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON Canada
| | - Tim Ramsay
- grid.28046.380000 0001 2182 2255School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON Canada
| | - Guy Trudel
- grid.412687.e0000 0000 9606 5108Department of Medicine, Division of Physical Medicine and Rehabilitation, Ottawa Hospital Research Institute, Ottawa, ON Canada ,grid.28046.380000 0001 2182 2255Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON Canada
| |
Collapse
|
39
|
Aaron N, Kraakman MJ, Zhou Q, Liu Q, Costa S, Yang J, Liu L, Yu L, Wang L, He Y, Fan L, Hirakawa H, Ding L, Lo J, Wang W, Zhao B, Guo E, Sun L, Rosen CJ, Qiang L. Adipsin promotes bone marrow adiposity by priming mesenchymal stem cells. eLife 2021; 10:69209. [PMID: 34155972 PMCID: PMC8219379 DOI: 10.7554/elife.69209] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/07/2021] [Indexed: 01/12/2023] Open
Abstract
Background Marrow adipose tissue (MAT) has been shown to be vital for regulating metabolism and maintaining skeletal homeostasis in the bone marrow (BM) niche. As a reflection of BM remodeling, MAT is highly responsive to nutrient fluctuations, hormonal changes, and metabolic disturbances such as obesity and diabetes mellitus. Expansion of MAT has also been strongly associated with bone loss in mice and humans. However, the regulation of BM plasticity remains poorly understood, as does the mechanism that links changes in marrow adiposity with bone remodeling. Methods We studied deletion of Adipsin, and its downstream effector, C3, in C57BL/6 mice as well as the bone-protected PPARγ constitutive deacetylation 2KR mice to assess BM plasticity. The mice were challenged with thiazolidinedione treatment, calorie restriction, or aging to induce bone loss and MAT expansion. Analysis of bone mineral density and marrow adiposity was performed using a μCT scanner and by RNA analysis to assess adipocyte and osteoblast markers. For in vitro studies, primary bone marrow stromal cells were isolated and subjected to osteoblastogenic or adipogenic differentiation or chemical treatment followed by morphological and molecular analyses. Clinical data was obtained from samples of a previous clinical trial of fasting and high-calorie diet in healthy human volunteers. Results We show that Adipsin is the most upregulated adipokine during MAT expansion in mice and humans in a PPARγ acetylation-dependent manner. Genetic ablation of Adipsin in mice specifically inhibited MAT expansion but not peripheral adipose depots, and improved bone mass during calorie restriction, thiazolidinedione treatment, and aging. These effects were mediated through its downstream effector, complement component C3, to prime common progenitor cells toward adipogenesis rather than osteoblastogenesis through inhibiting Wnt/β-catenin signaling. Conclusions Adipsin promotes new adipocyte formation and affects skeletal remodeling in the BM niche. Our study reveals a novel mechanism whereby the BM sustains its own plasticity through paracrine and endocrine actions of a unique adipokine. Funding This work was supported by the National Institutes of Health T32DK007328 (NA), F31DK124926 (NA), R01DK121140 (JCL), R01AR068970 (BZ), R01AR071463 (BZ), R01DK112943 (LQ), R24DK092759 (CJR), and P01HL087123 (LQ).
Collapse
Affiliation(s)
- Nicole Aaron
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pharmacology, Columbia UniversityNew YorkUnited States
| | - Michael J Kraakman
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Medicine, Columbia UniversityNew YorkUnited States
| | - Qiuzhong Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical SchoolSingaporeSingapore
| | - Qiongming Liu
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Samantha Costa
- Center for Molecular Medicine, Maine Medical Center Research InstituteScarboroughUnited States,School of Medicine, Tufts UniversityBostonUnited States,Graduate School of Biomedical Science and Engineering, University of MaineOronoUnited States
| | - Jing Yang
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Longhua Liu
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Lexiang Yu
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Liheng Wang
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Medicine, Columbia UniversityNew YorkUnited States
| | - Ying He
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Lihong Fan
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Hiroyuki Hirakawa
- Department of Microbiology and Immunology, Columbia UniversityNew YorkUnited States,Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - Lei Ding
- Department of Microbiology and Immunology, Columbia UniversityNew YorkUnited States,Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - James Lo
- Weill Center for Metabolic Health, Cardiovascular Research Institute, and Division of Cardiology, Weill Cornell Medical CollegeNew YorkUnited States
| | - Weidong Wang
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science CenterOklahoma CityUnited States
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and The David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, Department of Medicine, Weill Cornell Medical College; Graduate Program in Cell & Developmental Biology, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Edward Guo
- Department of Biomedical Engineering, Columbia UniversityNew YorkUnited States
| | - Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical SchoolSingaporeSingapore
| | - Cliff J Rosen
- Center for Molecular Medicine, Maine Medical Center Research InstituteScarboroughUnited States
| | - Li Qiang
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Epidemiologic studies reveal that the link between obesity and osteoarthritis cannot be uniquely explained by overweight-associated mechanical overload. For this reason, much attention focuses on the endocrine activity of adipose tissues. In addition to the systemic role of visceral and subcutaneous adipose tissues, many arguments highlight the involvement of local adipose tissues in osteoarthritis. RECENT FINDINGS Alteration in MRI signal intensity of the infrapatellar fat pad may predict both accelerated knee osteoarthritis and joint replacement. In this context, recent studies show that mesenchymal stromal cells could play a pivotal role in the pathological remodelling of intra-articular adipose tissues (IAATs) in osteoarthritis. In parallel, recent findings underline bone marrow adipose tissue as a major player in the control of the bone microenvironment, suggesting its possible role in osteoarthritis. SUMMARY The recent description of adipose tissues of various phenotypes within an osteoarthritic joint allows us to evoke their direct involvement in the initiation and progression of the osteoarthritic process. We can expect in the near future the discovery of novel molecules targeting these tissues.
Collapse
Affiliation(s)
| | - Florent Eymard
- Department of Rheumatology, AP-HP Henri Mondor Hospital
- Gly-CRRET Research Unit 4397, Université Paris-Est Créteil
| | - Francis Berenbaum
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA)
- Sorbonne Université, INSERM CRSA, AP-HP Hopital Saint Antoine, Paris, France
| | - Xavier Houard
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA)
| |
Collapse
|
41
|
Arai F, Stumpf PS, Ikushima YM, Hosokawa K, Roch A, Lutolf MP, Suda T, MacArthur BD. Machine Learning of Hematopoietic Stem Cell Divisions from Paired Daughter Cell Expression Profiles Reveals Effects of Aging on Self-Renewal. Cell Syst 2020; 11:640-652.e5. [DOI: 10.1016/j.cels.2020.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 05/22/2020] [Accepted: 11/10/2020] [Indexed: 12/30/2022]
|
42
|
Little-Letsinger SE, Pagnotti GM, McGrath C, Styner M. Exercise and Diet: Uncovering Prospective Mediators of Skeletal Fragility in Bone and Marrow Adipose Tissue. Curr Osteoporos Rep 2020; 18:774-789. [PMID: 33068251 PMCID: PMC7736569 DOI: 10.1007/s11914-020-00634-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW To highlight recent basic, translational, and clinical works demonstrating exercise and diet regulation of marrow adipose tissue (MAT) and bone and how this informs current understanding of the relationship between marrow adiposity and musculoskeletal health. RECENT FINDINGS Marrow adipocytes accumulate in the bone in the setting of not only hypercaloric intake (calorie excess; e.g., diet-induced obesity) but also with hypocaloric intake (calorie restriction; e.g., anorexia), despite the fact that these states affect bone differently. With hypercaloric intake, bone quantity is largely unaffected, whereas with hypocaloric intake, bone quantity and quality are greatly diminished. Voluntary running exercise in rodents was found to lower MAT and promote bone in eucaloric and hypercaloric states, while degrading bone in hypocaloric states, suggesting differential modulation of MAT and bone, dependent upon whole-body energy status. Energy status alters bone metabolism and bioenergetics via substrate availability or excess, which plays a key role in the response of bone and MAT to mechanical stimuli. Marrow adipose tissue (MAT) is a fat depot with a potential role in-as well as responsivity to-whole-body energy metabolism. Understanding the localized function of this depot in bone cell bioenergetics and substrate storage, principally in the exercised state, will aid to uncover putative therapeutic targets for skeletal fragility.
Collapse
Affiliation(s)
- Sarah E Little-Letsinger
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina, Chapel Hill, NC, USA.
| | - Gabriel M Pagnotti
- Department of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Cody McGrath
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Maya Styner
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
43
|
Zinngrebe J, Debatin KM, Fischer-Posovszky P. Adipocytes in hematopoiesis and acute leukemia: friends, enemies, or innocent bystanders? Leukemia 2020; 34:2305-2316. [PMID: 32474572 PMCID: PMC7449871 DOI: 10.1038/s41375-020-0886-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023]
Abstract
The bone marrow is home to well-balanced normal hematopoiesis, but also the stage of leukemia's crime. Marrow adipose tissue (MAT) is a unique and versatile component of the bone marrow niche. While the importance of MAT for bone health has long been recognized, its complex role in hematopoiesis has only recently gained attention. In this review article we summarize recent conceptual advances in the field of MAT research and how these developments impact our understanding of MAT regulation of hematopoiesis. Elucidating routes of interaction and regulation between MAT and cells of the hematopoietic system are essential to pinpoint vulnerable processes resulting in malignant transformation. The concept of white adipose tissue contributing to cancer development and progression on the cellular, metabolic, and systemic level is generally accepted. The role of MAT in malignant hematopoiesis, however, is controversial. MAT is very sensitive to changes in the patient's metabolic status hampering a clear definition of its role in different clinical situations. Here, we discuss future directions for leukemia research in the context of metabolism-induced modifications of MAT and other adipose tissues and how this might impact on leukemia cell survival, proliferation, and antileukemic therapy.
Collapse
Affiliation(s)
- Julia Zinngrebe
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Centre, D-89075, Ulm, Germany
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Centre, D-89075, Ulm, Germany
| | - Pamela Fischer-Posovszky
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Centre, D-89075, Ulm, Germany.
| |
Collapse
|
44
|
Li Z, MacDougald OA. Stem cell factor: the bridge between bone marrow adipocytes and hematopoietic cells. Haematologica 2020; 104:1689-1691. [PMID: 31473604 DOI: 10.3324/haematol.2019.224188] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Ziru Li
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ormond A MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
45
|
Abstract
Unhealthy diet, lack of exercise, psychosocial stress, and insufficient sleep are increasingly prevalent modifiable risk factors for cardiovascular disease. Accumulating evidence indicates that these risk factors may fuel chronic inflammatory processes that are active in atherosclerosis and lead to myocardial infarction and stroke. In concert with hyperlipidemia, maladaptive immune system activities can contribute to disease progression and increase the probability of adverse events. In this review, we discuss recent insight into how the above modifiable risk factors influence innate immunity. Specifically, we focus on pathways that raise systemic myeloid cell numbers and modulate immune cell phenotypes, reviewing hematopoiesis, leukocyte trafficking, and innate immune cell accumulation in cardiovascular organs. Often, relevant mechanisms that begin with lifestyle choices and lead to cardiovascular events span multiple organ systems, including the central nervous, endocrine, metabolic, hematopoietic, immune and, finally, the cardiovascular system. We argue that deciphering such pathways provides not only support for preventive interventions but also opportunities to develop biomimetic immunomodulatory therapeutics that mitigate cardiovascular inflammation.
Collapse
Affiliation(s)
- Maximilian J Schloss
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston (M.J.S., F.K.S., M.N.).,Department of Radiology, Massachusetts General Hospital, Boston (M.J.S., F.K.S., M.N.)
| | - Filip K Swirski
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston (M.J.S., F.K.S., M.N.).,Department of Radiology, Massachusetts General Hospital, Boston (M.J.S., F.K.S., M.N.)
| | - Matthias Nahrendorf
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston (M.J.S., F.K.S., M.N.).,Department of Radiology, Massachusetts General Hospital, Boston (M.J.S., F.K.S., M.N.).,Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.N.).,Department of Internal Medicine I, University Hospital Wuerzburg, Germany (M.N.)
| |
Collapse
|
46
|
Robino JJ, Pamir N, Rosario S, Crawford LB, Burwitz BJ, Roberts CT, Kurre P, Varlamov O. Spatial and biochemical interactions between bone marrow adipose tissue and hematopoietic stem and progenitor cells in rhesus macaques. Bone 2020; 133:115248. [PMID: 31972314 PMCID: PMC7085416 DOI: 10.1016/j.bone.2020.115248] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/11/2019] [Accepted: 01/18/2020] [Indexed: 01/11/2023]
Abstract
Recent developments in in situ microscopy have enabled unparalleled resolution of the architecture of the bone marrow (BM) niche for murine hematopoietic stem and progenitor cells (HSPCs). However, the extent to which these observations can be extrapolated to human BM remains unknown. In humans, adipose tissue occupies a significant portion of the BM medullary cavity, making quantitative immunofluorescent analysis difficult due to lipid-mediated light scattering. In this study, we employed optical clearing, confocal microscopy and nearest neighbor analysis to determine the spatial distribution of CD34+ HSPCs in the BM in a translationally relevant rhesus macaque model. Immunofluorescent analysis revealed that femoral BM adipocytes are associated with the branches of vascular sinusoids, with half of HSPCs localizing in close proximity of the nearest BM adipocyte. Immunofluorescent microscopy and flow cytometric analysis demonstrate that BM adipose tissue exists as a multicellular niche consisted of adipocytes, endothelial cells, granulocytes, and macrophages. Analysis of BM adipose tissue conditioned media using liquid chromatography-tandem mass spectrometry revealed the presence of multiple bioactive proteins involved in regulation of hematopoiesis, inflammation, and bone development, with many predicted to reside inside microvesicles. Pretreatment of purified HSPCs with BM adipose tissue conditioned media, comprising soluble and exosomal/microvesicle-derived factors, led to enhanced proliferation and an increase in granulocyte-monocyte differentiation potential ex vivo. Our work extends extensive studies in murine models, indicating that BM adipose tissue is a central paracrine regulator of hematopoiesis in nonhuman primates and possibly in humans.
Collapse
Affiliation(s)
- Jacob J Robino
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Nathalie Pamir
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Sara Rosario
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Lindsey B Crawford
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Benjamin J Burwitz
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA; Division of Pathobiology and Immunology, Oregon National Primate Center, USA
| | - Charles T Roberts
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR 97006, USA; Division of Reproductive and Developmental Sciences, Oregon National Primate Center, USA
| | - Peter Kurre
- Comprehensive Bone Marrow Failure Center, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Oleg Varlamov
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR 97006, USA.
| |
Collapse
|
47
|
Abstract
PURPOSE OF THE REVIEW The purpose of this review is to describe the in vitro and in vivo methods that researchers use to model and investigate bone marrow adipocytes (BMAds). RECENT FINDINGS The bone marrow (BM) niche is one of the most interesting and dynamic tissues of the human body. Relatively little is understood about BMAds, perhaps in part because these cells do not easily survive flow cytometry and histology processing and hence have been overlooked. Recently, researchers have developed in vitro and in vivo models to study normal function and dysfunction in the BM niche. Using these models, scientists and clinicians have noticed that BMAds, which form bone marrow adipose tissue (BMAT), are able to respond to numerous signals and stimuli, and communicate with local cells and distant tissues in the body. This review provides an overview of how BMAds are modeled and studied in vitro and in vivo.
Collapse
Affiliation(s)
- Michaela R Reagan
- Center for Molecular Medicine and Center for Translational Research, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA.
- University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA.
- School of Medicine and Graduate School of Biomedical Sciences, Tufts University, Boston, MA, USA.
| |
Collapse
|