553 results on '"Bone marrow niche"'
Search Results
2. Effect of bone marrow niche on hematopoietic stem cells.
- Author
-
Niazi, Vahid and Ghafouri-Fard, Soudeh
- Abstract
Hematopoietic stem cells (HSCs) reside in a milieu that supports their functions, differentiation, and survival. This niche consists of several types of cells, including mesenchymal stem/stromal cells, endothelial cells, osteoblasts, megakaryocytes, macrophages, adipocytes, lymphoid cells, and nerve fibers. The interactions between these cells and HSCs have a role in HSC fate. Several studies have focused on identification of the biological and cellular mechanisms contributing to the establishment of this niche. However, the exact mechanisms of the interaction between HSCs and the bone marrow niche have not been elucidated yet. Unraveling these mechanisms would help in the design of effective methods for maintenance and multiplication of HSCs in clinical settings, in addition to establishment of novel therapies for hematopoietic diseases. The current review summarizes the effects of the niche cells on HSC function and underlying mechanisms of interplay between HSCs and their niche. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
3. Advances in hematopoietic stem cells ex vivo expansion associated with bone marrow niche.
- Author
-
Deng, Ju, Tan, Yanhong, Xu, Zhifang, and Wang, Hongwei
- Abstract
Hematopoietic stem cells (HSCs) are an ideal source for the treatment of many hematological diseases and malignancies, as well as diseases of other systems, because of their two important features, self-renewal and multipotential differentiation, which have the ability to rebuild the blood system and immune system of the body. However, so far, the insufficient number of available HSCs, whether from bone marrow (BM), mobilized peripheral blood or umbilical cord blood, is still the main restricting factor for the clinical application. Therefore, strategies to expand HSCs numbers and maintain HSCs functions through ex vivo culture are urgently required. In this review, we outline the basic biology characteristics of HSCs, and focus on the regulatory factors in BM niche affecting the functions of HSCs. Then, we introduce several representative strategies used for HSCs from these three sources ex vivo expansion associated with BM niche. These findings have deepened our understanding of the mechanisms by which HSCs balance self-renewal and differentiation and provided a theoretical basis for the efficient clinical HSCs expansion. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Intraosseous Delivery of Mesenchymal Stem Cells for the Treatment of Bone and Hematological Diseases
- Author
-
Mikhail Yu. Artamonov and Evgeniy L. Sokov
- Subjects
mesenchymal stem cells (MSC) ,intraosseous delivery ,bone marrow niche ,tissue regeneration ,osteogenesis ,immunomodulation ,Biology (General) ,QH301-705.5 - Abstract
Mesenchymal stem cells are used most in regenerative medicine due to their capacities in differentiation and immune modulation. The intraosseous injection of MSC into the bone has been recommended because of expected outcomes for retention, bioavailability, and enhanced therapeutic efficacy, particularly in conditions involving the bone, such as osteoporosis and osteonecrosis. A review of the intraosseous delivery of mesenchymal stem cells in comparison with intravenous and intra-arterial delivery methods will be subjected to critical examination. This delivery mode fares better regarding paracrine signaling and immunomodulation attributes, which are the cornerstone of tissue regeneration and inflammation reduction. The local complications and technical challenges still apply with this method. This study was more focused on further research soon to be conducted to further elucidate long-term safety and efficacy of intraosseous mesenchymal stem cell therapy. Though much has been achieved with very impressive progress in this field, it is worth noting that more studies need to be put into place so that this technique can be established as a routine approach, especially with further research in biomaterials, gene therapy, and personalized medicine.
- Published
- 2024
- Full Text
- View/download PDF
5. Obesity modulates hematopoietic stem cell fate decision via IL-1β induced p38/MAPK signaling pathway
- Author
-
Jinxiao Yan, Pan Zhang, Xiru Liu, Chengwei Pan, Guolin Shi, Penghui Ye, Xiaohang Zou, Xiang Li, Xinmin Zheng, Yu Liu, and Hui Yang
- Subjects
Obesity ,Inflammation ,Hematopoietic stem cells ,Bone marrow niche ,IL-1β ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Obesity is accompanied by inflammation, which significantly affects the homeostasis of the immune microenvironment. Hematopoietic stem cells (HSCs), residing primarily in the bone marrow, play a vital role in maintaining and producing diverse mature blood cell lineages for the adult hematopoietic and immune systems. However, how HSCs development is affected by obese-promoting inflammation, and the mechanism by which HSC hematopoietic potency is affected by inflammatory signals originating from the obese-promoting changes on bone marrow niche remain unclear. This study elucidates the relationship between obesity-promoting inflammation and HSC fate determination. Methods The obesity mice model was established by feeding C57BL/6J mice a high-fat diet (HFD) containing 60% kcal fat. After 6 weeks, HSCs were analyzed using flow cytometry and identified key inflammation cytokine. Transcriptome sequencing techniques were used to discern the distinct pathways in HSCs. Ultimately, confirming the biological mechanism of obesity-induced HSC fate changes via Anakinra blocking specific inflammatory signals. Results Obesity caused by HFD changed the physical and biochemical properties of the bone marrow niche. In the HFD mice, the population of long-term HSCs in the bone marrow was decreased and facilitated HSCs differentiation towards the myeloid lineage. In addition, HFD increased expression of the inflammatory factor IL-1β in the bone marrow, and a significantly increased expression of IL-1r1 and active p38/MAPK signaling pathway were detected in the HSCs. Inhibition of IL-1β further normalized the expression of genes in p38/MAPK pathway and reversed HSC fate. Conclusions These findings have been demonstrated that the p38/MAPK signaling pathway in HSCs is activated by elevated levels of IL-1β within the HSC niche in obese models, thereby regulating HSC differentiation. It suggested a direct link between obesity-promoting inflammation and myeloid differentiation bias of HSCs in the HFD mice. Graphical abstract
- Published
- 2024
- Full Text
- View/download PDF
6. Hematopoietic and leukemic stem cells homeostasis: the role of bone marrow niche
- Author
-
Shaimaa Khattab, Manal El Sorady, Ashraf El-Ghandour, Giuseppe Visani, and Pier Paolo Piccaluga
- Subjects
hematopoietic stem cell ,acute myeloid leukemia ,bone marrow niche ,targeted therapy ,drug resistance ,precision medicine ,immune system ,Internal medicine ,RC31-1245 - Abstract
The bone marrow microenvironment (BMM) has highly specialized anatomical characteristics that provide a sanctuary place for hematopoietic stem cells (HSCs) that allow appropriate proliferation, maintenance, and self-renewal capacity. Several cell types contribute to the constitution and function of the bone marrow niche. Interestingly, uncovering the secrets of BMM and its interaction with HSCs in health paved the road for research aiming at better understanding the concept of leukemic stem cells (LSCs) and their altered niche. In fact, they share many signals that are responsible for interactions between LSCs and the bone marrow niche, due to several biological similarities between LSCs and HSCs. On the other hand, LSCs differ from HSCs in their abnormal activation of important signaling pathways that regulate survival, proliferation, drug resistance, invasion, and spread. Targeting these altered niches can help in better treatment choices for hematological malignancies and bone marrow disorders in general and acute myeloid leukemia (AML) in particular. Moreover, targeting those niches may help in decreasing the emergence of drug resistance and lower the relapse rate. In this article, the authors reviewed the most recent literature on bone marrow niches and their relations with either normal HSCs and AML cells/LSC, by focusing on pathogenetic and therapeutic implications.
- Published
- 2024
- Full Text
- View/download PDF
7. Obesity modulates hematopoietic stem cell fate decision via IL-1β induced p38/MAPK signaling pathway.
- Author
-
Yan, Jinxiao, Zhang, Pan, Liu, Xiru, Pan, Chengwei, Shi, Guolin, Ye, Penghui, Zou, Xiaohang, Li, Xiang, Zheng, Xinmin, Liu, Yu, and Yang, Hui
- Subjects
- *
HEMATOPOIETIC stem cells , *STEM cell niches , *HEMATOPOIETIC system , *BLOOD cells , *HIGH-fat diet - Abstract
Background: Obesity is accompanied by inflammation, which significantly affects the homeostasis of the immune microenvironment. Hematopoietic stem cells (HSCs), residing primarily in the bone marrow, play a vital role in maintaining and producing diverse mature blood cell lineages for the adult hematopoietic and immune systems. However, how HSCs development is affected by obese-promoting inflammation, and the mechanism by which HSC hematopoietic potency is affected by inflammatory signals originating from the obese-promoting changes on bone marrow niche remain unclear. This study elucidates the relationship between obesity-promoting inflammation and HSC fate determination. Methods: The obesity mice model was established by feeding C57BL/6J mice a high-fat diet (HFD) containing 60% kcal fat. After 6 weeks, HSCs were analyzed using flow cytometry and identified key inflammation cytokine. Transcriptome sequencing techniques were used to discern the distinct pathways in HSCs. Ultimately, confirming the biological mechanism of obesity-induced HSC fate changes via Anakinra blocking specific inflammatory signals. Results: Obesity caused by HFD changed the physical and biochemical properties of the bone marrow niche. In the HFD mice, the population of long-term HSCs in the bone marrow was decreased and facilitated HSCs differentiation towards the myeloid lineage. In addition, HFD increased expression of the inflammatory factor IL-1β in the bone marrow, and a significantly increased expression of IL-1r1 and active p38/MAPK signaling pathway were detected in the HSCs. Inhibition of IL-1β further normalized the expression of genes in p38/MAPK pathway and reversed HSC fate. Conclusions: These findings have been demonstrated that the p38/MAPK signaling pathway in HSCs is activated by elevated levels of IL-1β within the HSC niche in obese models, thereby regulating HSC differentiation. It suggested a direct link between obesity-promoting inflammation and myeloid differentiation bias of HSCs in the HFD mice. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Mechanobiology and Primary Cilium in the Pathophysiology of Bone Marrow Myeloproliferative Diseases.
- Author
-
Tiberio, Federica, Coda, Anna Rita Daniela, Tosi, Domiziano Dario, Luzi, Debora, Polito, Luca, Liso, Arcangelo, and Lattanzi, Wanda
- Subjects
- *
BONE marrow diseases , *BONE marrow cells , *STEM cell niches , *HEMATOPOIETIC stem cells , *POLYCYTHEMIA vera , *CILIA & ciliary motion - Abstract
Philadelphia-Negative Myeloproliferative neoplasms (MPNs) are a diverse group of blood cancers leading to excessive production of mature blood cells. These chronic diseases, including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), can significantly impact patient quality of life and are still incurable in the vast majority of the cases. This review examines the mechanobiology within a bone marrow niche, emphasizing the role of mechanical cues and the primary cilium in the pathophysiology of MPNs. It discusses the influence of extracellular matrix components, cell-cell and cell-matrix interactions, and mechanosensitive structures on hematopoietic stem cell (HSC) behavior and disease progression. Additionally, the potential implications of the primary cilium as a chemo- and mechanosensory organelle in bone marrow cells are explored, highlighting its involvement in signaling pathways crucial for hematopoietic regulation. This review proposes future research directions to better understand the dysregulated bone marrow niche in MPNs and to identify novel therapeutic targets. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Syndecans in hematopoietic cells and their niches.
- Author
-
Hagen, Matthew W., Setiawan, Nicollette J., Woodruff, Kelsey A., and Termini, Christina M.
- Subjects
- *
SYNDECANS , *HEPARAN sulfate proteoglycans , *HEMATOPOIETIC stem cells , *HEMATOPOIETIC system , *CELL adhesion , *SYSTEMS biology - Abstract
Heparan sulfate proteoglycans are a family of glycoproteins that modulate cell signaling by binding growth factors and changing their bioavailability. Syndecans are a specific family of transmembrane heparan sulfate proteoglycans that regulate cell adhesion, migration, and signaling. In this review, we will summarize emerging evidence for the functions of syndecans in the normal and malignant blood systems and their microenvironments. More specifically, we detail the known functions of syndecans within normal hematopoietic stem cells. Furthermore, we discuss the functions of syndecans in hematological malignancies, including myeloid malignancies, lymphomas, and bleeding disorders. As normal and malignant hematopoietic cells require cues from their microenvironments to function, we also summarize the roles of syndecans in cells of the stromal, endothelial, and osteolineage compartments. Syndecan biology is a rapidly evolving field; a comprehensive understanding of these molecules and their place in the hematopoietic system promises to improve our grasp on disease processes and better predict the efficacies of growth factor-targeting therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Ecological insights into hematopoiesis regulation: unraveling the influence of gut microbiota
- Author
-
Kaiwen Zheng, Zhifeng Wei, and Wei Li
- Subjects
Hematopoietic stem cell ,gut microbiota ,bone marrow niche ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
ABSTRACTThe gut microbiota constitutes a vast ecological system within the human body, forming a mutually interdependent entity with the host. In recent years, advancements in molecular biology technologies have provided a clearer understanding of the role of the gut microbiota. They not only influence the local immune status and metabolic functions of the host’s intestinal tract but also impact the functional transformation of hematopoietic stem cells (HSCs) through the gut-blood axis. In this review, we will discuss the role of the gut microbiota in influencing hematopoiesis. We analyze the interactions between HSCs and other cellular components, with a particular emphasis on the direct functional regulation of HSCs by the gut microbiota and their indirect influence through cellular components in the bone marrow microenvironment. Additionally, we propose potential control targets for signaling pathways triggered by the gut microbiota to regulate hematopoietic function, filling crucial knowledge gaps in the development of this research field.
- Published
- 2024
- Full Text
- View/download PDF
11. Cigarette smoke impairs the hematopoietic supportive property of mesenchymal stem cells via the production of reactive oxygen species and NLRP3 activation
- Author
-
Hyun Sung Park, Byung-Chul Lee, Dong-Hoon Chae, Aaron Yu, Jae Han Park, Jiyoung Heo, Myoung Hee Han, Keonwoo Cho, Joong Won Lee, Ji-Won Jung, Cynthia E. Dunbar, Mi-Kyung Oh, and Kyung-Rok Yu
- Subjects
Cigarette smoking extract ,Mesenchymal stem cells ,Reactive oxygen species ,NLRP3 ,Bone marrow niche ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Mesenchymal stem cells (MSCs) play important roles in tissue homeostasis by providing a supportive microenvironmental niche for the hematopoietic system. Cigarette smoking induces systemic abnormalities, including an impeded recovery process after hematopoietic stem cell transplantation. However, the role of cigarette smoking-mediated alterations in MSC niche function have not been investigated. Methods In the present study, we investigated whether exposure to cigarette smoking extract (CSE) disrupts the hematopoietic niche function of MSCs, and pathways impacted. To investigate the effects on bone marrow (BM)-derived MSCs and support of hematopoietic stem and progenitor cells (HSPCs), mice were repeatedly infused with the CSE named 3R4F, and hematopoietic stem and progenitor cells (HSPCs) supporting function was determined. The impact of 3R4F on MSCs at cellular level were screened by bulk-RNA sequencing and subsequently validated through qRT-PCR. Specific inhibitors were treated to verify the ROS or NLRP3-specific effects, and the cells were then transplanted into the animal model or subjected to coculture with HSPCs. Results Both direct ex vivo and systemic in vivo MSC exposure to 3R4F resulted in impaired engraftment in a humanized mouse model. Furthermore, transcriptomic profile analysis showed significantly upregulated signaling pathways related to reactive oxygen species (ROS), inflammation, and aging in 3R4F-treated MSCs. Notably, ingenuity pathway analysis revealed the activation of NLRP3 inflammasome signaling pathway in 3R4F-treated MSCs, and pretreatment with the NLRP3 inhibitor MCC950 rescued the HSPC-supporting ability of 3R4F-treated MSCs. Conclusion In conclusion, these findings indicate that exposure to CSE reduces HSPCs supportive function of MSCs by inducing robust ROS production and subsequent NLRP3 activation.
- Published
- 2024
- Full Text
- View/download PDF
12. Asparagine transport through SLC1A5/ASCT2 and SLC38A5/SNAT5 is essential for BCP‐ALL cell survival and a potential therapeutic target.
- Author
-
Taurino, Giuseppe, Dander, Erica, Chiu, Martina, Pozzi, Giulia, Maccari, Chiara, Starace, Rita, Silvestri, Daniela, Griffini, Erika, Bianchi, Massimiliano G., Carubbi, Cecilia, Andreoli, Roberta, Mirandola, Prisco, Valsecchi, Maria Grazia, Rizzari, Carmelo, D'Amico, Giovanna, and Bussolati, Ovidio
- Subjects
- *
CELL survival , *ASPARAGINE , *LYMPHOBLASTIC leukemia , *ACUTE leukemia , *STROMAL cells , *BLAST injuries - Abstract
Summary: B‐cell precursor acute lymphoblastic leukaemia (BCP‐ALL) blasts strictly depend on the transport of extra‐cellular asparagine (Asn), yielding a rationale for L‐asparaginase (ASNase) therapy. However, the carriers used by ALL blasts for Asn transport have not been identified yet. Exploiting RS4;11 cells as BCP‐ALL model, we have found that cell Asn is lowered by either silencing or inhibition of the transporters ASCT2 or SNAT5. The inhibitors V‐9302 (for ASCT2) and GluγHA (for SNAT5) markedly lower cell proliferation and, when used together, suppress mTOR activity, induce autophagy and cause a severe nutritional stress, leading to a proliferative arrest and a massive cell death in both the ASNase‐sensitive RS4;11 cells and the relatively ASNase‐insensitive NALM‐6 cells. The cytotoxic effect is not prevented by coculturing leukaemic cells with primary mesenchymal stromal cells. Leukaemic blasts of paediatric ALL patients express ASCT2 and SNAT5 at diagnosis and undergo marked cytotoxicity when exposed to the inhibitors. ASCT2 expression is positively correlated with the minimal residual disease at the end of the induction therapy. In conclusion, ASCT2 and SNAT5 are the carriers exploited by ALL cells to transport Asn, and ASCT2 expression is associated with a lower therapeutic response. ASCT2 may thus represent a novel therapeutic target in BCP‐ALL. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Acute malaria suppresses the B lymphocytic niche in the bone marrow through the alteration of CXCL12-abundant reticular cells.
- Author
-
Lee, Michelle Sue Jann, Matsuo-Dapaah, Julia, Zorrilla, Camila Del Rosario, Omatsu, Yoshiki, Nagasawa, Takashi, Uemura, Shun, Iwama, Atsushi, Ishii, Ken J, and Coban, Cevayir
- Subjects
- *
BONE marrow , *BONE marrow cells , *MESENCHYMAL stem cells , *MALARIA , *HEMATOPOIETIC stem cells - Abstract
Bone marrow is a dynamic organ composed of stem cells that constantly receive signals from stromal cells and other hematopoietic cells in the niches of the bone marrow to maintain hematopoiesis and generate immune cells. Perturbation of the bone marrow microenvironment by infection and inflammation affects hematopoiesis and may affect immune cell development. Little is known about the effect of malaria on the bone marrow stromal cells that govern the hematopoietic stem cell (HSC) niche. In this study, we demonstrate that the mesenchymal stromal CXCL12-abundant reticular (CAR) cell population is reduced during acute malaria infection. The reduction of CXCL12 and interleukin-7 signals in the bone marrow impairs the lymphopoietic niche, leading to the depletion of common lymphoid progenitors, B cell progenitors, and mature B cells, including plasma cells in the bone marrow. We found that interferon-γ (IFNγ) is responsible for the upregulation of Sca1 on CAR cells, yet the decline in CAR cell and B cell populations in the bone marrow is IFNγ-independent. In contrast to the decline in B cell populations, HSCs and multipotent progenitors increased with the expansion of myelopoiesis and erythropoiesis, indicating a bias in the differentiation of multipotent progenitors during malaria infection. These findings suggest that malaria may affect host immunity by modulating the bone marrow niche. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Cigarette smoke impairs the hematopoietic supportive property of mesenchymal stem cells via the production of reactive oxygen species and NLRP3 activation.
- Author
-
Park, Hyun Sung, Lee, Byung-Chul, Chae, Dong-Hoon, Yu, Aaron, Park, Jae Han, Heo, Jiyoung, Han, Myoung Hee, Cho, Keonwoo, Lee, Joong Won, Jung, Ji-Won, Dunbar, Cynthia E., Oh, Mi-Kyung, and Yu, Kyung-Rok
- Subjects
- *
CIGARETTE smoke , *REACTIVE oxygen species , *MESENCHYMAL stem cells , *SMOKING , *NLRP3 protein , *HEMATOPOIETIC stem cell transplantation - Abstract
Background: Mesenchymal stem cells (MSCs) play important roles in tissue homeostasis by providing a supportive microenvironmental niche for the hematopoietic system. Cigarette smoking induces systemic abnormalities, including an impeded recovery process after hematopoietic stem cell transplantation. However, the role of cigarette smoking-mediated alterations in MSC niche function have not been investigated. Methods: In the present study, we investigated whether exposure to cigarette smoking extract (CSE) disrupts the hematopoietic niche function of MSCs, and pathways impacted. To investigate the effects on bone marrow (BM)-derived MSCs and support of hematopoietic stem and progenitor cells (HSPCs), mice were repeatedly infused with the CSE named 3R4F, and hematopoietic stem and progenitor cells (HSPCs) supporting function was determined. The impact of 3R4F on MSCs at cellular level were screened by bulk-RNA sequencing and subsequently validated through qRT-PCR. Specific inhibitors were treated to verify the ROS or NLRP3-specific effects, and the cells were then transplanted into the animal model or subjected to coculture with HSPCs. Results: Both direct ex vivo and systemic in vivo MSC exposure to 3R4F resulted in impaired engraftment in a humanized mouse model. Furthermore, transcriptomic profile analysis showed significantly upregulated signaling pathways related to reactive oxygen species (ROS), inflammation, and aging in 3R4F-treated MSCs. Notably, ingenuity pathway analysis revealed the activation of NLRP3 inflammasome signaling pathway in 3R4F-treated MSCs, and pretreatment with the NLRP3 inhibitor MCC950 rescued the HSPC-supporting ability of 3R4F-treated MSCs. Conclusion: In conclusion, these findings indicate that exposure to CSE reduces HSPCs supportive function of MSCs by inducing robust ROS production and subsequent NLRP3 activation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Complementary and Inducible creERT2 Mouse Models for Functional Evaluation of Endothelial Cell Subtypes in the Bone Marrow.
- Author
-
Poulos, Michael G., Ramalingam, Pradeep, Winiarski, Agatha, Gutkin, Michael C., Katsnelson, Lizabeth, Carter, Cody, Pibouin-Fragner, Laurence, Eichmann, Anne, Thomas, Jean-Leon, Miquerol, Lucile, and Butler, Jason M.
- Subjects
- *
BONE marrow cells , *ENDOTHELIAL cells , *HEMATOPOIETIC stem cells , *BONE marrow , *LABORATORY mice , *ANIMAL disease models - Abstract
In the adult bone marrow (BM), endothelial cells (ECs) are an integral component of the hematopoietic stem cell (HSC)-supportive niche, which modulates HSC activity by producing secreted and membrane-bound paracrine signals. Within the BM, distinct vascular arteriole, transitional, and sinusoidal EC subtypes display unique paracrine expression profiles and create anatomically-discrete microenvironments. However, the relative contributions of vascular endothelial subtypes in supporting hematopoiesis is unclear. Moreover, constitutive expression and off-target activity of currently available endothelial-specific and endothelial-subtype-specific murine cre lines potentially confound data analysis and interpretation. To address this, we describe two tamoxifen-inducible cre-expressing lines, Vegfr3-creERT2 and Cx40-creERT2, that efficiently label sinusoidal/transitional and arteriole endothelium respectively in adult marrow, without off-target activity in hematopoietic or perivascular cells. Utilizing an established mouse model in which cre-dependent recombination constitutively-activates MAPK signaling within adult endothelium, we identify arteriole ECs as the driver of MAPK-mediated hematopoietic dysfunction. These results define complementary tamoxifen-inducible creERT2-expressing mouse lines that label functionally-discrete and non-overlapping sinusoidal/transitional and arteriole EC populations in the adult BM, providing a robust toolset to investigate the differential contributions of vascular subtypes in maintaining hematopoietic homeostasis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. Immunomodulatory properties of CD38 antibodies and their effect on anticancer efficacy in multiple myeloma
- Author
-
Kamlesh Bisht, Taro Fukao, Marielle Chiron, Paul Richardson, Djordje Atanackovic, Eduardo Chini, Wee Joo Chng, Helgi Van De Velde, and Fabio Malavasi
- Subjects
adenosine ,bone marrow niche ,CD38 antibodies ,daratumumab ,immunomodulation ,isatuximab ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background CD38 has been established as an important therapeutic target for multiple myeloma (MM), for which two CD38 antibodies are currently approved—daratumumab and isatuximab. CD38 is an ectoenzyme that degrades NAD and its precursors and is involved in the production of adenosine and other metabolites. Aim Among the various mechanisms by which CD38 antibodies can induce MM cell death is immunomodulation, including multiple pathways for CD38‐mediated T‐cell activation. Patients who respond to anti‐CD38 targeting treatment experience more marked changes in T‐cell expansion, activity, and clonality than nonresponders. Implications Resistance mechanisms that undermine the immunomodulatory effects of CD38‐targeting therapies can be tumor intrinsic, such as the downregulation of CD38 surface expression and expression of complement inhibitor proteins, and immune microenvironment‐related, such as changes to the natural killer (NK) cell numbers and function in the bone marrow niche. There are numerous strategies to overcome this resistance, which include identifying and targeting other therapeutic targets involved in, for example, adenosine production, the activation of NK cells or monocytes through immunomodulatory drugs and their combination with elotuzumab, or with bispecific T‐cell engagers.
- Published
- 2023
- Full Text
- View/download PDF
17. Abnormal adipogenic signaling in the bone marrow mesenchymal stem cells contributes to supportive microenvironment for leukemia development
- Author
-
Rawan Sabbah, Sahar Saadi, Tal Shahar-Gabay, Shiran Gerassy, Shlomit Yehudai-Resheff, and Tsila Zuckerman
- Subjects
Mesenchymal stem cells ,Bone marrow niche ,Acute myeloid leukemia ,Adipocytes ,Osteocytes ,Medicine ,Cytology ,QH573-671 - Abstract
Abstract Background Acute myeloid leukemia (AML) is an aggressive hematological malignancy, associated with unfavorable patient outcome, primarily due to disease relapse. Mesenchymal stem cells (MSCs) residing in the bone marrow (BM) niche are the source of mesenchyma-derived subpopulations, including adipocytes, and osteocytes, that are critical for normal hematopoiesis. This study aimed to characterize BM-derived adipocyte/osteocyte fractions and their crosstalk with AML cells as a potential mechanism underlying leukemogenesis. Methods BM cell subpopulations derived from primary AML patients were evaluated using humanized ex-vivo and in-vivo models, established for this study. The models comprised AML blasts, normal hematopoietic stem and progenitor cells and mesenchymal stromal subpopulations. ELISA, FACS analysis, colony forming unit assay, whole exome sequencing and real-time qPCR were employed to assess the differentiation capacity, genetic status, gene expression and function of these cell fractions. To explore communication pathways between AML cells and BM subpopulations, levels of signaling mediators, including cytokines and chemokines, were measured using the ProcartaPlex multiplex immunoassay. Results The study revealed deficiencies in adipogenic/osteogenic differentiation of BM-MSCs derived from AML patients, with adipocytes directly promoting survival and clonogenicity of AML cells in-vitro. In whole exome sequencing of BM-MSC/stromal cells, the AHNAK2 gene, associated with the stimulation of adipocyte differentiation, was found to be mutated and significantly under-expressed, implying its abnormal function in AML. The evaluation of communication pathways between AML cells and BM subpopulations demonstrated pronounced alterations in the crosstalk between these cell fractions. This was reflected by significantly elevated levels of signaling mediators cytokines/chemokines, in AML-induced adipocytes/osteocytes compared to non-induced MSCs, indicating abnormal hematopoiesis. Furthermore, in-vivo experiments using a fully humanized 3D scaffold model, showed that AML-induced adipocytes were the dominant component of the tumor microenvironment, providing preferential support to leukemia cell survival and proliferation. Conclusions This study has disclosed direct contribution of impaired functional, genetic and molecular properties of AML patient-derived adipocytes to effective protection of AML blasts from apoptosis and to stimulation of their growth in vitro and in vivo, which overall leads to disease propagation and relapse. The detected AHNAK2 gene mutations in AML-MSCs point to their involvement in the mechanism underlying abnormal adipogenesis. Video Abstract
- Published
- 2023
- Full Text
- View/download PDF
18. Exploring the intricate cross-talk between clonal expansion and the bone marrow niche
- Author
-
Steven Ngo, Despoina Papazoglou, Hector Huerga Encabo, and Dominique Bonnet
- Subjects
bone marrow niche ,myeloid leukaemia ,clonal haematopoiesis ,microenvironment ,stromal cells ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Haematopoietic stem cells (HSCs) reside within an intricate network of cells in the bone marrow (BM) niche. HSC crosstalk with niche compartments influences lineage determination and blood cell production, while independent niche interactions are essential for the maintenance of HSC quiescence. How different niche components influence the genetic diversity of HSCs represents an expanding field of investigation. As such, we will summarise the current knowledge of the contribution to the Darwinian evolution of mutant HSCs of both haematopoietic and non-haematopoietic cells residing in the BM. In this review, we will disentangle how somatic evolution associates with the niche at two stages: from (1) the stage of preleukaemic HSC expansion and clonal haematopoiesis (CH) to (2) leukaemia-initiating cells (LICs) and the development of myeloid malignancies with acute myeloid leukaemia (AML) being the most prevalent. We will finally describe current challenges such as limitations in models used in the field or the difficulty in studying specific genetic clones in isolation.
- Published
- 2024
- Full Text
- View/download PDF
19. Complementary and Inducible creERT2 Mouse Models for Functional Evaluation of Endothelial Cell Subtypes in the Bone Marrow
- Author
-
Poulos, Michael G., Ramalingam, Pradeep, Winiarski, Agatha, Gutkin, Michael C., Katsnelson, Lizabeth, Carter, Cody, Pibouin-Fragner, Laurence, Eichmann, Anne, Thomas, Jean-Leon, Miquerol, Lucile, and Butler, Jason M.
- Published
- 2024
- Full Text
- View/download PDF
20. Immunomodulatory properties of CD38 antibodies and their effect on anticancer efficacy in multiple myeloma.
- Author
-
Bisht, Kamlesh, Fukao, Taro, Chiron, Marielle, Richardson, Paul, Atanackovic, Djordje, Chini, Eduardo, Chng, Wee Joo, Van De Velde, Helgi, and Malavasi, Fabio
- Subjects
- *
CD38 antigen , *MULTIPLE myeloma , *ANTINEOPLASTIC agents , *KILLER cells , *IMMUNOGLOBULINS , *ADENOSINES , *ECULIZUMAB , *ACTIVATED protein C resistance - Abstract
Background: CD38 has been established as an important therapeutic target for multiple myeloma (MM), for which two CD38 antibodies are currently approved—daratumumab and isatuximab. CD38 is an ectoenzyme that degrades NAD and its precursors and is involved in the production of adenosine and other metabolites. Aim: Among the various mechanisms by which CD38 antibodies can induce MM cell death is immunomodulation, including multiple pathways for CD38‐mediated T‐cell activation. Patients who respond to anti‐CD38 targeting treatment experience more marked changes in T‐cell expansion, activity, and clonality than nonresponders. Implications: Resistance mechanisms that undermine the immunomodulatory effects of CD38‐targeting therapies can be tumor intrinsic, such as the downregulation of CD38 surface expression and expression of complement inhibitor proteins, and immune microenvironment‐related, such as changes to the natural killer (NK) cell numbers and function in the bone marrow niche. There are numerous strategies to overcome this resistance, which include identifying and targeting other therapeutic targets involved in, for example, adenosine production, the activation of NK cells or monocytes through immunomodulatory drugs and their combination with elotuzumab, or with bispecific T‐cell engagers. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
21. Abnormal adipogenic signaling in the bone marrow mesenchymal stem cells contributes to supportive microenvironment for leukemia development.
- Author
-
Sabbah, Rawan, Saadi, Sahar, Shahar-Gabay, Tal, Gerassy, Shiran, Yehudai-Resheff, Shlomit, and Zuckerman, Tsila
- Subjects
- *
ADIPOGENESIS , *MESENCHYMAL stem cells , *COLONY-forming units assay , *BONE marrow , *HEMATOPOIETIC stem cells , *ACUTE myeloid leukemia - Abstract
Background: Acute myeloid leukemia (AML) is an aggressive hematological malignancy, associated with unfavorable patient outcome, primarily due to disease relapse. Mesenchymal stem cells (MSCs) residing in the bone marrow (BM) niche are the source of mesenchyma-derived subpopulations, including adipocytes, and osteocytes, that are critical for normal hematopoiesis. This study aimed to characterize BM-derived adipocyte/osteocyte fractions and their crosstalk with AML cells as a potential mechanism underlying leukemogenesis. Methods: BM cell subpopulations derived from primary AML patients were evaluated using humanized ex-vivo and in-vivo models, established for this study. The models comprised AML blasts, normal hematopoietic stem and progenitor cells and mesenchymal stromal subpopulations. ELISA, FACS analysis, colony forming unit assay, whole exome sequencing and real-time qPCR were employed to assess the differentiation capacity, genetic status, gene expression and function of these cell fractions. To explore communication pathways between AML cells and BM subpopulations, levels of signaling mediators, including cytokines and chemokines, were measured using the ProcartaPlex multiplex immunoassay. Results: The study revealed deficiencies in adipogenic/osteogenic differentiation of BM-MSCs derived from AML patients, with adipocytes directly promoting survival and clonogenicity of AML cells in-vitro. In whole exome sequencing of BM-MSC/stromal cells, the AHNAK2 gene, associated with the stimulation of adipocyte differentiation, was found to be mutated and significantly under-expressed, implying its abnormal function in AML. The evaluation of communication pathways between AML cells and BM subpopulations demonstrated pronounced alterations in the crosstalk between these cell fractions. This was reflected by significantly elevated levels of signaling mediators cytokines/chemokines, in AML-induced adipocytes/osteocytes compared to non-induced MSCs, indicating abnormal hematopoiesis. Furthermore, in-vivo experiments using a fully humanized 3D scaffold model, showed that AML-induced adipocytes were the dominant component of the tumor microenvironment, providing preferential support to leukemia cell survival and proliferation. Conclusions: This study has disclosed direct contribution of impaired functional, genetic and molecular properties of AML patient-derived adipocytes to effective protection of AML blasts from apoptosis and to stimulation of their growth in vitro and in vivo, which overall leads to disease propagation and relapse. The detected AHNAK2 gene mutations in AML-MSCs point to their involvement in the mechanism underlying abnormal adipogenesis. AaFEVfq6u-1YqH7bnQ85ua Video Abstract [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
22. Cytotoxic Conditioning-Induced Changes in Bone Marrow Microenvironment: A Study in Wild-Type and AML Mouse Models
- Author
-
ABBASIZADEH, NASTARAN
- Subjects
Biology ,Immunology ,Acute Myeloid Leukemia ,Bone Marrow Niche ,Chemotherapy ,Cytotoxic Conditioning Intensity ,Recipient Age - Abstract
Hematopoiesis depends on complex interactions between hematopoietic stem cells (HSCs) and the bone marrow (BM) microenvironment. However, alterations in this regulated system can lead to malignant transformation and hematopoietic diseases. Acute myeloid leukemia (AML) is characterized by uncontrolled growth of leukemic blasts in the BM and is the most common acute leukemia in adults. Tumor survival after cytotoxic treatment of AML patients remains a major therapeutic challenge, contributing to disease relapse. Fine-tuning the cytotoxic conditioning regimen to discover the most effective treatment plan has the potential to significantly improve outcomes in AML patients, thereby reducing the risk of relapse. The mechanism by which conditioning achieves therapeutic outcomes is through BM ablation. Additionally, conditioning can impact different compartments of the BM microenvironment. In this study, we investigated the impact of busulfan conditioning on the BM niche, focusing on how the intensity of cytotoxic conditioning regimens and animal age influence this dynamic process. We later expanded our findings to an AML mouse model to evaluate the BM niche around resistant tumor cells after cytotoxic therapy.By examining the impact of varying dosages and recipient age factors on treatment response as well as BM microenvironment adjacent to the residual tumor cells after therapy, we sought to optimize chemotherapy regimens and establish the groundwork for tailoring treatment strategies to AML cancer patients.
- Published
- 2024
23. Consequences of HIV infection in the bone marrow niche.
- Author
-
Herd, Candice Lee, Mellet, Juanita, Mashingaidze, Tsungai, Durandt, Chrisna, and Pepper, Michael Sean
- Subjects
HIV infections ,BONE marrow ,BONE marrow cells ,BLOOD cells ,CELL communication - Abstract
Dysregulation of the bone marrow niche resulting from the direct and indirect effects of HIV infection contributes to haematological abnormalities observed in HIV patients. The bone marrow niche is a complex, multicellular environment which functions primarily in the maintenance of haematopoietic stem/progenitor cells (HSPCs). These adult stem cells are responsible for replacing blood and immune cells over the course of a lifetime. Cells of the bone marrow niche support HSPCs and help to orchestrate the quiescence, self-renewal and differentiation of HSPCs through chemical and molecular signals and cell-cell interactions. This narrative review discusses the HIV-associated dysregulation of the bone marrow niche, as well as the susceptibility of HSPCs to infection by HIV. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
24. Bone marrow microenvironment disruption and sustained inflammation with prolonged haematologic toxicity after CAR T‐cell therapy.
- Author
-
Kitamura, Wataru, Asada, Noboru, Naoi, Yusuke, Abe, Masaya, Fujiwara, Hideaki, Ennishi, Daisuke, Nishimori, Hisakazu, Fujii, Keiko, Fujii, Nobuharu, Matsuoka, Ken‐ichi, Yoshino, Tadashi, and Maeda, Yoshinobu
- Subjects
- *
BONE marrow , *DIFFUSE large B-cell lymphomas , *T cells , *STEM cell factor , *CHIMERIC antigen receptors - Abstract
Summary: Mechanisms of prolonged cytopenia (PC) after chimeric antigen receptor (CAR) T‐cell therapy, an emerging therapy for relapsed or refractory diffuse large B‐cell lymphoma, remain elusive. Haematopoiesis is tightly regulated by the bone marrow (BM) microenvironment, called the 'niche'. To investigate whether alterations in the BM niche cells are associated with PC, we analysed CD271+ stromal cells in BM biopsy specimens and the cytokine profiles of the BM and serum obtained before and on day 28 after CAR T‐cell infusion. Imaging analyses of the BM biopsy specimens revealed that CD271+ niche cells were severely impaired after CAR T‐cell infusion in patients with PC. Cytokine analyses after CAR T‐cell infusion showed that CXC chemokine ligand 12 and stem cell factor, niche factors essential for haematopoietic recovery, were significantly decreased in the BM of patients with PC, suggesting reduced niche cell function. The levels of inflammation‐related cytokines on day 28 after CAR T‐cell infusion were consistently high in the BM of patients with PC. Thus, we demonstrate for the first time that BM niche disruption and sustained elevation of inflammation‐related cytokines in the BM following CAR T‐cell infusion are associated with subsequent PC. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
25. Electrical Sympathetic Neuromodulation Protects Bone Marrow Niche and Drives Hematopoietic Regeneration during Chemotherapy.
- Author
-
Hsu, Ya‐Ting, Chen, Li‐Hsien, Liu, Ya‐Hui, Chu, Shih‐Kai, Chen, Tsai‐Yun, Tsai, Kuen‐Jer, Shen, Meng‐Ru, and Liu, Wentai
- Subjects
- *
BONE marrow , *GRANULOCYTE-colony stimulating factor , *STEM cell factor , *PERIPHERAL nerve injuries , *HEMATOPOIETIC stem cells , *SYMPATHETIC nervous system - Abstract
The sympathetic nervous system (SNS) of the bone marrow regulates the regeneration and mobilization of hematopoietic stem cells. Chemotherapy can damage bone marrow SNS, which impairs hematopoietic regeneration and aggravates hematologic toxicities. This leads to long‐term bone marrow niche damage and increases mortality in patients undergoing chemotherapy. Electrical neuromodulation has been used to improve functional recovery after peripheral nerve injury. This study demonstrates that electrical sympathetic neuromodulation (ESN) of bone marrow can protect the bone marrow niche from chemotherapy‐induced injury. Using carboplatin‐treated rats, the SNS via the sciatic nerve innervating the femoral marrow with the effective protocol for bone marrow sympathetic activation is electrically stimulated. ESN can mediate several hematopoietic stem cells maintenance factors and promote hematopoietic regeneration after chemotherapy. It also activates adrenergic signals and reduces the release of pro‐inflammatory cytokines, particularly interleukin‐1 β, which contribute to chemotherapy‐related nerve injury. Consequently, the severity of chemotherapy‐related leukopenia, thrombocytopenia, and mortality can be reduced by ESN. As a result, in contrast to current drug‐based treatment, such as granulocyte colony‐stimulating factor, ESN can be a disruptive adjuvant treatment by protecting and modulating bone marrow function to reduce hematologic toxicity during chemotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
26. Diagnostic utility of mast cell analysis in hypoplastic bone marrows for differentiation of aplastic anaemia from hypoplastic myelodysplastic neoplasm: A retrospective study [version 1; peer review: 2 not approved]
- Author
-
Chaithra G V, Soumya Gupta, Ranjitha Rao, Sharada Rai, Nirupama Murali, and Sunita Hegde
- Subjects
Research Article ,Articles ,Mast cell ,Aplastic anaemia ,MDS-h ,Hypocellular marrow ,Bone marrow niche ,CD34 ,Hematopoietic stem cell. - Abstract
Background: Haematological disorders characterized by hypocellular marrow like aplastic anaemia (AA) and hypocellular myelodysplastic neoplasm (MDS-h) are diagnostic challenges as the clinical and morphological features overlap, while the treatment and prognosis differ. Bone marrow mast cell quantification is proposed as a supplementary diagnostic and prognostic tool to differentiate AA from MDS-h. We aim to study the distribution of mast cells in hypoplastic marrow and determine its role in differential diagnosis of AA and MDS-h. Methods: We analysed bone marrow biopsies of 65 cases of hypoplastic/aplastic marrow received during the year 2015-2021 for the distribution of mast cells using Toluidine Blue special staining. Hematopoietic stem cells were assessed with the help of CD34 immunohistochemistry. Results: Increased mast cells were noted in 55.4% of all the cases, of which 48% were cases of AA and 21% were cases of MDS-h (P: 0.04). Overall, 76% of AA and 33% of MDS-h cases showed increased numbers of mast cells. Reduced hematopoietic stem cells were noted in 69.4% of the cases with elevated mast cells (P: 0.1). Conclusions: Mast cell quantification in trephine biopsy of AA and MDS-h can act as a supplementary diagnostic tool and guide the management of the respective entities. Understanding its role in the bone marrow niche can enhance stem cell transplant treatment.
- Published
- 2023
- Full Text
- View/download PDF
27. Osteoblasts and osteoclasts: an important switch of tumour cell dormancy during bone metastasis
- Author
-
Rongchen Dai, Mengfan Liu, Xincheng Xiang, Zhichao Xi, and Hongxi Xu
- Subjects
Tumour cell dormancy ,Osteoclasts ,Osteoblasts ,Bone marrow niche ,Bone metastasis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Bone metastasis occurs when tumour cells dissociate from primary tumours, enter the circulation (circulating tumour cells, CTCs), and colonize sites in bone (disseminated tumour cells, DTCs). The bone marrow seems to be a particularly dormancy-inducing environment for DTCs, yet the mechanisms of dormancy initiation, reactivation, and interaction within the bone marrow have to be elucidated. Intriguingly, some evidence has suggested that dormancy is a reversible state that is switched ‘on’ or ‘off’ depending on the presence of various bone marrow resident cells, particularly osteoclasts and osteoblasts. It has become clear that these two cells contribute to regulating dormant tumour cells in bone both directly (interaction) and indirectly (secreted factors). The involved mechanisms include TGFβ signalling, the Wnt signalling axis, the Notch2 pathway, etc. There is no detailed review that specifically focuses on ascertaining the dynamic interactions between tumour cell dormancy and bone remodelling. In addition, we highlighted the roles of inflammatory cytokines during this ‘cell-to-cell’ communication. We also discussed the potential clinical relevance of remodelling the bone marrow niche in controlling dormant tumour cells. Understanding the unique role of osteoclasts and osteoblasts in regulating tumour dormancy in bone marrow will provide new insight into preventing and treating tumour bone metastasis.
- Published
- 2022
- Full Text
- View/download PDF
28. Consequences of HIV infection in the bone marrow niche
- Author
-
Candice Lee Herd, Juanita Mellet, Tsungai Mashingaidze, Chrisna Durandt, and Michael Sean Pepper
- Subjects
bone marrow niche ,HIV ,haematopoiesis ,haematopoietic stem/progenitor cell ,dysregulation ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Dysregulation of the bone marrow niche resulting from the direct and indirect effects of HIV infection contributes to haematological abnormalities observed in HIV patients. The bone marrow niche is a complex, multicellular environment which functions primarily in the maintenance of haematopoietic stem/progenitor cells (HSPCs). These adult stem cells are responsible for replacing blood and immune cells over the course of a lifetime. Cells of the bone marrow niche support HSPCs and help to orchestrate the quiescence, self-renewal and differentiation of HSPCs through chemical and molecular signals and cell-cell interactions. This narrative review discusses the HIV-associated dysregulation of the bone marrow niche, as well as the susceptibility of HSPCs to infection by HIV.
- Published
- 2023
- Full Text
- View/download PDF
29. Extracellular Vesicles and MicroRNA in Myelodysplastic Syndromes.
- Author
-
Meunier, Mathieu, Laurin, David, and Park, Sophie
- Subjects
- *
EXTRACELLULAR vesicles , *MYELODYSPLASTIC syndromes , *MICRORNA , *BONE marrow , *STROMAL cells , *EXOSOMES , *HEMATOPOIETIC stem cells - Abstract
The bone marrow niche plays an increasing role in the pathophysiogenesis of myelodysplastic syndromes. More specifically, mesenchymal stromal cells, which can secrete extracellular vesicles and their miRNA contents, modulate the fate of hematopoietic stem cells leading to leukemogenesis. Extracellular vesicles can mediate their miRNA and protein contents between nearby cells but also in the plasma of the patients, being potent tools for diagnosis and prognostic markers in MDS. They can be targeted by antisense miRNA or by modulators of the secretion of extracellular vesicles and could lead to future therapeutic directions in MDS. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
30. ERRα is dispensable for hematopoietic stem cell function in the bone marrow.
- Author
-
Bae, Hee Eun, Baryawno, Ninib, Kim, Byoung Soo, Yoon, Sik, Oh, Sae-Ock, Jo, Eun-Kyeong, Kim, Chi Dae, and Lee, Dongjun
- Subjects
- *
HEMATOPOIETIC stem cells , *BONE marrow cells , *BLOOD cells , *ACUTE myeloid leukemia , *BONE marrow - Abstract
Hematopoietic stem cells (HSCs) have the ability to self-renew, differentiate into various blood cell types, and reside in the bone marrow (BM) niche. Estrogen-related receptor α (ERRα) is an orphan nuclear receptor that has a role in mitochondrial biogenesis and metabolic regulation. Previous research has shown that ERRα contributes to the development of acute myeloid leukemia (AML) by acting as a key regulator of mitochondrial processes, though its role in HSC regulation remains mostly unknown. Flow cytometric analysis determined the population of the hematopoietic stem and progenitor cells (HSPC) using Errα knockout (KO) and conditional KO (cKO) mice. Furthermore, we investigated reconstitution with BM transplantation assay. Here, the ablation of Errα in the BM demonstrated that the production of mature blood cells, lineage distribution within hematopoietic organs, and frequencies of the HSPC populations were similar to those of controls. In addition, the ablation of Errα did not perturb HSC function under the stress of transplantation. Collectively, ERRα is not necessary for the control of HSPC populations, as well as for the maintenance of HSC characteristics and functions within the BM. • ERRα deletion does not affect HSC function or lineage distribution in the bone marrow. • Hematopoietic stem and progenitor cell (HSPC) populations remain stable without ERRα. • ERRα deficiency does not impair HSC reconstitution under transplantation stress. • ERRα is dispensable for maintaining HSPC populations and HSC functions in the BM niche. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. Polyclonal regeneration of mouse bone marrow endothelial cells after irradiative conditioning.
- Author
-
Skulimowska, Izabella, Morys, Jan, Sosniak, Justyna, Gonka, Monika, Gulati, Gunsagar, Sinha, Rahul, Kowalski, Kacper, Mosiolek, Sylwester, Weissman, Irving L., Jozkowicz, Alicja, Szade, Agata, and Szade, Krzysztof
- Abstract
Bone marrow endothelial cells (BM-ECs) are the essential components of the BM niche and support the function of hematopoietic stem cells (HSCs). However, conditioning for HSC transplantation causes damage to the recipients' BM-ECs and may lead to transplantation-related morbidity. Here, we investigated the cellular and clonal mechanisms of BM-EC regeneration after irradiative conditioning. Using single-cell RNA sequencing, imaging, and flow cytometry, we revealed how the heterogeneous pool of BM-ECs changes during regeneration from irradiation stress. Next, we developed a single-cell in vitro clonogenic assay and demonstrated that all EC fractions hold a high potential to reenter the cell cycle and form vessel-like structures. Finally, we used Rainbow mice and a machine-learning-based model to show that the regeneration of BM-ECs after irradiation is mostly polyclonal and driven by the broad fraction of BM-ECs; however, the cell output among clones varies at later stages of regeneration. [Display omitted] • Bone marrow endothelial cells (BM-ECs) have high potential to reenter cell cycle • Regeneration of BM-ECs after irradiation is highly polyclonal • scRNA-seq reveals dynamic remodeling of BM-ECs after irradiation Irradiation, used for conditioning recipients before bone marrow (BM) transplantation, damages BM endothelial cells (BM-ECs). Skulimowska et al. elucidated the mechanisms behind BM-EC regeneration following irradiative conditioning. Their study revealed that a diverse population of BM-ECs can reenter the cell cycle and that the regeneration process is highly polyclonal. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Editorial: The role of the bone marrow microenvironment in multiple myeloma evolution and therapy
- Author
-
Carolina Schinke, Niels Weinhold, and Jesus Delgado-Calle
- Subjects
multiple myeloma ,bone marrow microenviroment ,bone marrow niche ,bone metabolism ,mesenchymal stem cells ,SPARC ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
33. Identification of the Human Skeletal Stem Cell
- Author
-
Chan, Charles KF, Gulati, Gunsagar S, Sinha, Rahul, Tompkins, Justin Vincent, Lopez, Michael, Carter, Ava C, Ransom, Ryan C, Reinisch, Andreas, Wearda, Taylor, Murphy, Matthew, Brewer, Rachel E, Koepke, Lauren S, Marecic, Owen, Manjunath, Anoop, Seo, Eun Young, Leavitt, Tripp, Lu, Wan-Jin, Nguyen, Allison, Conley, Stephanie D, Salhotra, Ankit, Ambrosi, Thomas H, Borrelli, Mimi R, Siebel, Taylor, Chan, Karen, Schallmoser, Katharina, Seita, Jun, Sahoo, Debashis, Goodnough, Henry, Bishop, Julius, Gardner, Michael, Majeti, Ravindra, Wan, Derrick C, Goodman, Stuart, Weissman, Irving L, Chang, Howard Y, and Longaker, Michael T
- Subjects
Medical Biotechnology ,Biomedical and Clinical Sciences ,Regenerative Medicine ,Genetics ,Stem Cell Research - Nonembryonic - Non-Human ,Clinical Research ,Stem Cell Research - Nonembryonic - Human ,Stem Cell Research ,Stem Cell Research - Induced Pluripotent Stem Cell ,Transplantation ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,1.1 Normal biological development and functioning ,Musculoskeletal ,Animals ,Bone Development ,Bone and Bones ,Cartilage ,Cell Differentiation ,Hematopoietic Stem Cells ,Humans ,Induced Pluripotent Stem Cells ,Mesenchymal Stem Cells ,Mice ,Mice ,Inbred C57BL ,Signal Transduction ,Single-Cell Analysis ,Stem Cells ,Stromal Cells ,Transcriptome ,ATAC-sequencing ,HSC ,and stromal progenitor ,bone ,bone fracture repair ,bone marrow niche ,cartilage ,human skeletal stem cell ,single cell RNA-sequencing ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Stem cell regulation and hierarchical organization of human skeletal progenitors remain largely unexplored. Here, we report the isolation of a self-renewing and multipotent human skeletal stem cell (hSSC) that generates progenitors of bone, cartilage, and stroma, but not fat. Self-renewing and multipotent hSSCs are present in fetal and adult bones and can also be derived from BMP2-treated human adipose stroma (B-HAS) and induced pluripotent stem cells (iPSCs). Gene expression analysis of individual hSSCs reveals overall similarity between hSSCs obtained from different sources and partially explains skewed differentiation toward cartilage in fetal and iPSC-derived hSSCs. hSSCs undergo local expansion in response to acute skeletal injury. In addition, hSSC-derived stroma can maintain human hematopoietic stem cells (hHSCs) in serum-free culture conditions. Finally, we combine gene expression and epigenetic data of mouse skeletal stem cells (mSSCs) and hSSCs to identify evolutionarily conserved and divergent pathways driving SSC-mediated skeletogenesis. VIDEO ABSTRACT.
- Published
- 2018
34. The biology of hematopoietic stem cells and its clinical implications.
- Author
-
Skulimowska, Izabella, Sosniak, Justyna, Gonka, Monika, Szade, Agata, Jozkowicz, Alicja, and Szade, Krzysztof
- Subjects
- *
BIOLOGY , *BONE marrow , *HEMATOPOIESIS , *BLOOD grouping & crossmatching , *CLINICAL medicine - Abstract
Hematopoietic stem cells (HSCs) give rise to all types of blood cells and self‐renew their own population. The regeneration potential of HSCs has already been successfully translated into clinical applications. However, recent studies on the biology of HSCs may further extend their clinical use in future. The roles of HSCs in native hematopoiesis and in transplantation settings may differ. Furthermore, the heterogenic pool of HSCs dynamically changes during aging. These changes also involve the complex interactions of HSCs with the bone marrow niche. Here, we review the opportunities and challenges of these findings to improve the clinical use of HSCs. We describe new methods of HSCs mobilization and conditioning for the transplantation of HSCs. Finally, we highlight the research findings that may lead to overcoming the current limitations of HSC transplantation and broaden the patient group that can benefit from the clinical potential of HSCs. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
35. Diversity in the bone marrow niche: Classic and novel strategies to uncover niche composition.
- Author
-
Sánchez‐Lanzas, Raúl, Kalampalika, Foteini, and Ganuza, Miguel
- Subjects
- *
BONE marrow , *HEMATOPOIETIC stem cells , *MULTIPOTENT stem cells , *CELL anatomy , *STROMAL cells - Abstract
Summary: Our view on the role and composition of the bone marrow (BM) has dramatically changed over time from a simple nutrient for the bone to a highly complex multicellular tissue that sustains haematopoiesis. Among these cells, multipotent haematopoietic stem cells (HSCs), which are predominantly quiescent, possess unique self‐renewal capacity and multilineage differentiation potential and replenish all blood lineages to maintain lifelong haematopoiesis. Adult HSCs reside in specialised BM niches, which support their functions. Much effort has been put into deciphering HSC niches due to their potential clinical relevance. Multiple cell types have been implicated as HSC‐niche components including sinusoidal endothelium, perivascular stromal cells, macrophages, megakaryocytes, osteoblasts and sympathetic nerves. In this review we provide a historical perspective on how technical advances, from genetic mouse models to imaging and high‐throughput sequencing techniques, are unveiling the plethora of molecular cues and cellular components that shape the niche and regulate HSC functions. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
36. Effect of bone marrow niche on hematopoietic stem cells.
- Author
-
Niazi V and Ghafouri-Fard S
- Subjects
- Humans, Animals, Cell Differentiation, Bone Marrow metabolism, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells cytology, Stem Cell Niche
- Abstract
Hematopoietic stem cells (HSCs) reside in a milieu that supports their functions, differentiation, and survival. This niche consists of several types of cells, including mesenchymal stem/stromal cells, endothelial cells, osteoblasts, megakaryocytes, macrophages, adipocytes, lymphoid cells, and nerve fibers. The interactions between these cells and HSCs have a role in HSC fate. Several studies have focused on identification of the biological and cellular mechanisms contributing to the establishment of this niche. However, the exact mechanisms of the interaction between HSCs and the bone marrow niche have not been elucidated yet. Unraveling these mechanisms would help in the design of effective methods for maintenance and multiplication of HSCs in clinical settings, in addition to establishment of novel therapies for hematopoietic diseases. The current review summarizes the effects of the niche cells on HSC function and underlying mechanisms of interplay between HSCs and their niche., Competing Interests: Declarations. Conflict of interest: The authors declare no competing interests. Ethical approval and consent to participate: Not applicable. Consent for publication: Not applicable., (© 2024. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
37. Intraosseous Delivery of Mesenchymal Stem Cells for the Treatment of Bone and Hematological Diseases.
- Author
-
Artamonov MY and Sokov EL
- Abstract
Mesenchymal stem cells are used most in regenerative medicine due to their capacities in differentiation and immune modulation. The intraosseous injection of MSC into the bone has been recommended because of expected outcomes for retention, bioavailability, and enhanced therapeutic efficacy, particularly in conditions involving the bone, such as osteoporosis and osteonecrosis. A review of the intraosseous delivery of mesenchymal stem cells in comparison with intravenous and intra-arterial delivery methods will be subjected to critical examination. This delivery mode fares better regarding paracrine signaling and immunomodulation attributes, which are the cornerstone of tissue regeneration and inflammation reduction. The local complications and technical challenges still apply with this method. This study was more focused on further research soon to be conducted to further elucidate long-term safety and efficacy of intraosseous mesenchymal stem cell therapy. Though much has been achieved with very impressive progress in this field, it is worth noting that more studies need to be put into place so that this technique can be established as a routine approach, especially with further research in biomaterials, gene therapy, and personalized medicine.
- Published
- 2024
- Full Text
- View/download PDF
38. c-Kit M541L variant is related to ineffective hemopoiesis predisposing to clonal evolution in 3D in vitro biomimetic co-culture model of bone marrow niche
- Author
-
Paola Manzo, Pasqualina Scala, Valentina Giudice, Marisa Gorrese, Angela Bertolini, Denise Morini, Francesca D'Alto, Rita Pepe, Antonio Pedicini, Barbara Izzo, Francesco Verdesca, Maddalena Langella, Bianca Serio, Giovanna Della Porta, and Carmine Selleri
- Subjects
3D cultures ,Bone marrow niche ,Clonal evolution ,Hematopoiesis ,Science (General) ,Q1-390 ,Social sciences (General) ,H1-99 - Abstract
Hematopoietic stem cell (HSC) maintenance in vitro is challenging because stem cell survival relies on cell-to-cell contacts and paracrine signals from bone marrow (BM) microenvironment. Indeed, HSCs easily differentiate in conventional culture systems, and in vitro study of stem cell biology, leukemogenesis, and evolutionary trajectories is limited. 3D-culture systems can mimic tissue architecture and microenvironment thus preserving HSC phenotype. In this study, we developed a calcium alginate hydrogel-based 3D co-culture system of BM mononuclear cells (BMMCs) and BM-derived mesenchymal stem cells (BM-MSCs) to study hemopoiesis in health and disease, such as biological roles of c-Kit M541L somatic mutation of unknown significance. BMMCs and peripheral blood stem cells were obtained from an acute myeloid leukemia patient who experienced graft failure and his haploidentical donor, and from a healthy donor. Cells embedded in alginate scaffolds were cultured for up to 21 days, and flow cytometry immunophenotyping was performed at baseline and every seven days. Our results showed suitability of our 3D culture system in preserving HSC vitality and phenotype throughout the culture period, and also in maintaining composition and vitality of total BMMCs. Moreover, 3D in vitro culture results suggested that M541L c-Kit somatic mutation could be a loss-of-function alteration by reducing HSC maintenance ability thus quickly promoting differentiation, as documented by in vivo graft failure and in vitro absence of long-term culture stability. In conclusions, our 3D BM-like biomimetic culture system allowed long-term stemness maintenance, making it a valid and effective tool for in vitro study of physiological and pathological hemopoiesis.
- Published
- 2022
- Full Text
- View/download PDF
39. Osteoblasts and osteoclasts: an important switch of tumour cell dormancy during bone metastasis.
- Author
-
Dai, Rongchen, Liu, Mengfan, Xiang, Xincheng, Xi, Zhichao, and Xu, Hongxi
- Subjects
- *
BONE metastasis , *BONE marrow , *OSTEOBLASTS , *OSTEOCLASTS , *BONE marrow cells , *BONE remodeling , *BONE cells - Abstract
Bone metastasis occurs when tumour cells dissociate from primary tumours, enter the circulation (circulating tumour cells, CTCs), and colonize sites in bone (disseminated tumour cells, DTCs). The bone marrow seems to be a particularly dormancy-inducing environment for DTCs, yet the mechanisms of dormancy initiation, reactivation, and interaction within the bone marrow have to be elucidated. Intriguingly, some evidence has suggested that dormancy is a reversible state that is switched 'on' or 'off' depending on the presence of various bone marrow resident cells, particularly osteoclasts and osteoblasts. It has become clear that these two cells contribute to regulating dormant tumour cells in bone both directly (interaction) and indirectly (secreted factors). The involved mechanisms include TGFβ signalling, the Wnt signalling axis, the Notch2 pathway, etc. There is no detailed review that specifically focuses on ascertaining the dynamic interactions between tumour cell dormancy and bone remodelling. In addition, we highlighted the roles of inflammatory cytokines during this 'cell-to-cell' communication. We also discussed the potential clinical relevance of remodelling the bone marrow niche in controlling dormant tumour cells. Understanding the unique role of osteoclasts and osteoblasts in regulating tumour dormancy in bone marrow will provide new insight into preventing and treating tumour bone metastasis. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
40. Perturbations of mesenchymal stromal cells after allogeneic hematopoietic cell transplantation predispose for bone marrow graft-versus-host-disease.
- Author
-
Krüger, Thomas, Wehner, Rebekka, Herbig, Maik, Kräter, Martin, Kramer, Michael, Middeke, Jan Moritz, Stölzel, Friedrich, List, Catrin, Egger-Heidrich, Katharina, Teipel, Raphael, Oelschlägel, Uta, Wermke, Martin, Jambor, Helena, Wobus, Manja, Schetelig, Johannes, Jöhrens, Korinna, Tonn, Torsten, Subburayalu, Julien, and von Bonin, Malte
- Subjects
HEMATOPOIETIC stem cell transplantation ,BONE marrow transplantation ,STROMAL cells ,BONE marrow ,GRAFT versus host disease - Abstract
Functional impairment of the bone marrow (BM) niche has been suggested as a major reason for prolonged cytopenia and secondary graft failure after allogeneic hematopoietic cell transplantation (alloHCT). Because mesenchymal stromal cells (MSCs) serve as multipotent progenitors for several niche components in the BM, they might play a key role in this process. We used collagenase digested trephine biopsies to directly quantify MSCs in 73 patients before (n = 18) and/or after alloHCT (n = 65). For the first time, we demonstrate that acute graft-versus-host disease (aGvHD, n = 39) is associated with a significant decrease in MSC numbers. MSC reduction can be observed even before the clinical onset of aGvHD (n = 10). Assessing MSCs instantly after biopsy collection revealed phenotypic and functional differences depending on the occurrence of aGvHD. These differences vanished during ex vivo expansion. The MSC endotypes observed revealed an enhanced population of donor-derived classical dendritic cells type 1 and alloreactive T cells as the causing agent for compartmental inflammation and MSC damage before clinical onset of aGvHD was ascertained. In conclusion, MSCs endotypes may constitute a predisposing conductor of alloreactivity after alloHCT preceding the clinical diagnosis of aGvHD. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
41. Identification of the Factor That Leads Human Mesenchymal Stem Cell Lines into Decellularized Bone.
- Author
-
Koyanagi, Anri, Onishi, Iichiroh, Muraoka, Karin, Sato, Ikue, Sato, Shingo, Kimura, Tsuyoshi, Kishida, Akio, Yamamoto, Kouhei, Kitagawa, Masanobu, and Kurata, Morito
- Subjects
- *
MESENCHYMAL stem cells , *HUMAN stem cells , *HEMATOPOIETIC stem cells , *CELL lines , *BONE marrow , *CELL migration - Abstract
Hematopoiesis is maintained by the interaction of hematopoietic stem cells (HSCs) and bone marrow mesenchymal stem cells (MSCs) in bone marrow microenvironments, called niches. Certain genetic mutations in MSCs, not HSCs, provoke some hematopoietic neoplasms, such as myelodysplastic syndrome. An in vivo bone marrow niche model using human MSC cell lines with specific genetic mutations and bone scaffolds is necessary to elucidate these interactions and the disease onset. We focused on decellularized bone (DCB) as a useful bone scaffold and attempted to induce human MSCs (UE7T-9 cells) into the DCB. Using the CRISPR activation library, we identified SHC4 upregulation as a candidate factor, with the SHC4 overexpression in UE7T-9 cells activating their migratory ability and upregulating genes to promote hematopoietic cell migration. This is the first study to apply the CRISPR library to engraft cells into decellularized biomaterials. SHC4 overexpression is essential for engrafting UE7T-9 cells into DCB, and it might be the first step toward creating an in vivo human–mouse hybrid bone marrow niche model. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
42. Comparative analysis of extracellular vesicle isolation methods from human AML bone marrow cells and AML cell lines.
- Author
-
Lang, Jonas B., Buck, Michèle C., Rivière, Jennifer, Stambouli, Oumaima, Sachenbacher, Ken, Choudhary, Purva, Dietz, Hendrik, Giebel, Bernd, Bassermann, Florian, Oostendorp, Robert A. J., Götze, Katharina S., and Hecker, Judith S.
- Subjects
BONE marrow cells ,EXTRACELLULAR vesicles ,GREEN fluorescent protein ,ACUTE myeloid leukemia ,CELL lines - Abstract
Cellular crosstalk between hematopoietic stem/progenitor cells and the bone marrow (BM) niche is vital for the development and maintenance of myeloid malignancies. These compartments can communicate via bidirectional transfer of extracellular vesicles (EVs). EV trafficking in acute myeloid leukemia (AML) plays a crucial role in shaping the BM microenvironment into a leukemia- permissive niche. Although several EV isolation methods have been developed, it remains a major challenge to define the most accurate and reliable procedure. Here, we tested the efficacy and functional assay compatibility of four different EV isolation methods in leukemia-derived EVs: (1) membrane affinity-based: exoEasy Kit alone and (2) in combination with Amicon filtration; (3) precipitation: ExoQuick-TC; and (4) ultracentrifugation (UC). Western blot analysis of EV fractions showed the highest enrichment of EV marker expression (e.g., CD63, HSP70, and TSG101) by precipitation with removal of overabundant soluble proteins [e.g., bovine serum albumin (BSA)], which were not discarded using UC. Besides the presence of damaged EVs after UC, intact EVs were successfully isolated with all methods as evidenced by highly maintainedspherical-andcup-shapedvesiclesintransmissionelectron microscopy. Nanoparticle tracking analysis of EV particle size and concentration revealed significant differences in EV isolation efficacy, with exoEasy Kit providing the highest EV yield recovery. Of note, functional assays with exoEasy Kit-isolated EVs showed significant toxicity towards treated target cells [e.g., mesenchymal stromal cells (MSCs)], which was abrogated when combining exoEasy Kit with Amicon filtration. Additionally, MSC treated with green fluorescent protein (GFP)-tagged exoEasy Kit-isolated EVs did not show any EV uptake, while EV isolation by precipitation demonstrated efficient EV internalization. Taken together, the choice of EV isolation procedure significantly impacts the yield and potential functionality of leukemia-derived EVs. The cheapest method (UC) resulted in contaminated and destructed EV fractions, while the isolation method with the highest EV yield (exoEasy Kit) appeared to be incompatible with functional assays. We identified two methods (precipitation-based ExoQuick-TC and membrane affinity-based exoEasy Kit combined with Amicon filtration) yielding pure and intact EVs, also suitable for application in functional assays. This study highlights the importance of selecting the right EV isolation method depending on the desired experimental design. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
43. Circular RNAs Activity in the Leukemic Bone Marrow Microenvironment.
- Author
-
Liccardo, Francesca, Iaiza, Alessia, Śniegocka, Martyna, Masciarelli, Silvia, and Fazi, Francesco
- Subjects
- *
CIRCULAR RNA , *BONE marrow , *HEMATOPOIETIC stem cells , *BONE marrow cells , *ACUTE myeloid leukemia , *HEMATOLOGIC malignancies - Abstract
Acute myeloid leukemia (AML) is a hematological malignancy originating from defective hematopoietic stem cells in the bone marrow. In spite of the recent approval of several molecular targeted therapies for AML treatment, disease recurrence remains an issue. Interestingly, increasing evidence has pointed out the relevance of bone marrow (BM) niche remodeling during leukemia onset and progression. Complex crosstalk between AML cells and microenvironment components shapes the leukemic BM niche, consequently affecting therapy responsiveness. Notably, circular RNAs are a new class of RNAs found to be relevant in AML progression and chemoresistance. In this review, we provided an overview of AML-driven niche remodeling. In particular, we analyzed the role of circRNAs and their possible contribution to cell–cell communication within the leukemic BM microenvironment. Understanding these mechanisms will help develop a more effective treatment for AML. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
44. Comparative analysis of extracellular vesicle isolation methods from human AML bone marrow cells and AML cell lines
- Author
-
Jonas B. Lang, Michèle C. Buck, Jennifer Rivière, Oumaima Stambouli, Ken Sachenbacher, Purva Choudhary, Hendrik Dietz, Bernd Giebel, Florian Bassermann, Robert A. J. Oostendorp, Katharina S. Götze, and Judith S. Hecker
- Subjects
extracellular vesicles ,intercellular communication ,AML ,bone marrow niche ,EV isolation methods ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Cellular crosstalk between hematopoietic stem/progenitor cells and the bone marrow (BM) niche is vital for the development and maintenance of myeloid malignancies. These compartments can communicate via bidirectional transfer of extracellular vesicles (EVs). EV trafficking in acute myeloid leukemia (AML) plays a crucial role in shaping the BM microenvironment into a leukemia-permissive niche. Although several EV isolation methods have been developed, it remains a major challenge to define the most accurate and reliable procedure. Here, we tested the efficacy and functional assay compatibility of four different EV isolation methods in leukemia-derived EVs: (1) membrane affinity-based: exoEasy Kit alone and (2) in combination with Amicon filtration; (3) precipitation: ExoQuick-TC; and (4) ultracentrifugation (UC). Western blot analysis of EV fractions showed the highest enrichment of EV marker expression (e.g., CD63, HSP70, and TSG101) by precipitation with removal of overabundant soluble proteins [e.g., bovine serum albumin (BSA)], which were not discarded using UC. Besides the presence of damaged EVs after UC, intact EVs were successfully isolated with all methods as evidenced by highly maintained spherical- and cup-shaped vesicles in transmission electron microscopy. Nanoparticle tracking analysis of EV particle size and concentration revealed significant differences in EV isolation efficacy, with exoEasy Kit providing the highest EV yield recovery. Of note, functional assays with exoEasy Kit-isolated EVs showed significant toxicity towards treated target cells [e.g., mesenchymal stromal cells (MSCs)], which was abrogated when combining exoEasy Kit with Amicon filtration. Additionally, MSC treated with green fluorescent protein (GFP)-tagged exoEasy Kit-isolated EVs did not show any EV uptake, while EV isolation by precipitation demonstrated efficient EV internalization. Taken together, the choice of EV isolation procedure significantly impacts the yield and potential functionality of leukemia-derived EVs. The cheapest method (UC) resulted in contaminated and destructed EV fractions, while the isolation method with the highest EV yield (exoEasy Kit) appeared to be incompatible with functional assays. We identified two methods (precipitation-based ExoQuick-TC and membrane affinity-based exoEasy Kit combined with Amicon filtration) yielding pure and intact EVs, also suitable for application in functional assays. This study highlights the importance of selecting the right EV isolation method depending on the desired experimental design.
- Published
- 2022
- Full Text
- View/download PDF
45. Perturbations of mesenchymal stromal cells after allogeneic hematopoietic cell transplantation predispose for bone marrow graft-versus-host-disease
- Author
-
Thomas Krüger, Rebekka Wehner, Maik Herbig, Martin Kräter, Michael Kramer, Jan Moritz Middeke, Friedrich Stölzel, Catrin List, Katharina Egger-Heidrich, Raphael Teipel, Uta Oelschlägel, Martin Wermke, Helena Jambor, Manja Wobus, Johannes Schetelig, Korinna Jöhrens, Torsten Tonn, Julien Subburayalu, Marc Schmitz, Martin Bornhauser, and Malte von Bonin
- Subjects
mesenchymal stromal cells ,allogeneic hematopoietic stem cell transplantation ,graft-versus-host-disease ,alloreactivity ,bone marrow niche ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Functional impairment of the bone marrow (BM) niche has been suggested as a major reason for prolonged cytopenia and secondary graft failure after allogeneic hematopoietic cell transplantation (alloHCT). Because mesenchymal stromal cells (MSCs) serve as multipotent progenitors for several niche components in the BM, they might play a key role in this process. We used collagenase digested trephine biopsies to directly quantify MSCs in 73 patients before (n = 18) and/or after alloHCT (n = 65). For the first time, we demonstrate that acute graft-versus-host disease (aGvHD, n = 39) is associated with a significant decrease in MSC numbers. MSC reduction can be observed even before the clinical onset of aGvHD (n = 10). Assessing MSCs instantly after biopsy collection revealed phenotypic and functional differences depending on the occurrence of aGvHD. These differences vanished during ex vivo expansion. The MSC endotypes observed revealed an enhanced population of donor-derived classical dendritic cells type 1 and alloreactive T cells as the causing agent for compartmental inflammation and MSC damage before clinical onset of aGvHD was ascertained. In conclusion, MSCs endotypes may constitute a predisposing conductor of alloreactivity after alloHCT preceding the clinical diagnosis of aGvHD.
- Published
- 2022
- Full Text
- View/download PDF
46. Neurological Regulation of the Bone Marrow Niche
- Author
-
Aerts-Kaya, Fatima, Ulum, Baris, Mammadova, Aynura, Köse, Sevil, Aydin, Gözde, Korkusuz, Petek, Uçkan-Çetinkaya, Duygu, Crusio, Wim E., Series Editor, Lambris, John D., Series Editor, Rezaei, Nima, Series Editor, and Turksen, Kursad, editor
- Published
- 2020
- Full Text
- View/download PDF
47. Heat shock protein and antioxidant enzyme gene expression and fold changes in bone marrow samples from healthy rats.
- Author
-
Mustafa, Walaa Aladdin and Alchalabi, Ali Saeed
- Subjects
HEAT shock proteins ,GENE expression ,BONE marrow ,HEMATOPOIESIS ,PROTEIN folding ,LABORATORY rats - Abstract
Copyright of Journal of Education & Science is the property of Republic of Iraq Ministry of Higher Education & Scientific Research (MOHESR) and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2022
- Full Text
- View/download PDF
48. Acute Myeloid Leukaemia Drives Metabolic Changes in the Bone Marrow Niche.
- Author
-
Maynard, Rebecca S., Hellmich, Charlotte, Bowles, Kristian M., and Rushworth, Stuart A.
- Subjects
ACUTE myeloid leukemia ,BONE marrow ,HEMATOPOIETIC stem cells ,BONE marrow cells ,OXIDATIVE phosphorylation - Abstract
Acute myeloid leukaemia (AML) is a highly proliferative cancer characterised by infiltration of immature haematopoietic cells in the bone marrow (BM). AML predominantly affects older people and outcomes, particularly in this difficult to treat population remain poor, in part due to inadequate response to therapy, and treatment toxicity. Normal haematopoiesis is supported by numerous support cells within the BM microenvironment or niche, including adipocytes, stromal cells and endothelial cells. In steady state haematopoiesis, haematopoietic stem cells (HSCs) primarily acquire ATP through glycolysis. However, during stress-responses HSCs rapidly transition to oxidative phosphorylation, enabled by mitochondrial plasticity. Historically it was thought that cancer cells preferentially used glycolysis for ATP production, however recently it has become evident that many cancers, including AML primarily use the TCA cycle and oxidative phosphorylation for rapid proliferation. AML cells hijack the stress-response pathways of their non-malignant counterparts, utilising mitochondrial changes to drive expansion. In addition, amino acids are also utilised by leukaemic stem cells to aid their metabolic output. Together, these processes allow AML cells to maximise their ATP production, using multiple metabolites and fuelling rapid cell turnover which is a hallmark of the disease. This review of AML derived changes in the BM niche, which enable enhanced metabolism, will consider the important pathways and discuss future challenges with a view to understanding how AML cells are able to hijack metabolic pathways and how we may elucidate new targets for potential therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
49. Inhibiting PI3K–AKT–mTOR Signaling in Multiple Myeloma-Associated Mesenchymal Stem Cells Impedes the Proliferation of Multiple Myeloma Cells.
- Author
-
Heinemann, Luca, Möllers, Klara Maria, Ahmed, Helal Mohammed Mohammed, Wei, Lanying, Sun, Kaiyan, Nimmagadda, Subbaiah Chary, Frank, Daria, Baumann, Anja, Poos, Alexandra M., Dugas, Martin, Varghese, Julian, Raab, Marc-Steffen, and Khandanpour, Cyrus
- Subjects
MESENCHYMAL stem cells ,MULTIPLE myeloma ,BONE marrow cancer ,CELL proliferation ,PROGNOSIS ,PLASMACYTOMA ,BORTEZOMIB ,PLASMA cell diseases - Abstract
The microenvironment of cancer cells is receiving increasing attention as an important factor influencing the progression and prognosis of tumor diseases. In multiple myeloma (MM), a hematological cancer of plasma cells, mesenchymal stem cells (MSCs) represent an integral part of the bone marrow niche and tumor microenvironment. It has been described that MM cells alter MSCs in a way that MM-associated MSCs promote the proliferation and survival of MM cells. Yet, our understanding of the molecular mechanisms governing the interaction between MM cells and MSCs and whether this can be targeted for therapeutic interventions is limited. To identify potential molecular targets, we examined MSCs by RNA sequencing and Western blot analysis. We report that MSCs from MM patients with active disease (MM-Act-MSCs) show a distinct gene expression profile as compared with MSCs from patients with other (non-) malignant diseases (CTR-MSCs). Of note, we detected a significant enrichment of the PI3K–AKT–mTOR hallmark gene set in MM-Act-MSCs and further confirmed the increased levels of related proteins in these MSCs. Pictilisib, a pan-PI3K inhibitor, selectively reduced the proliferation of MM-Act-MSCs as compared with CTR-MSCs. Furthermore, pictilisib treatment impaired the MM-promoting function of MM-Act-MSCs. Our data thus provide a deeper insight into the molecular signature and function of MSCs associated with MM and show that targeting PI3K–AKT–mTOR signaling in MSCs may represent an additional therapeutic pathway in the treatment of MM patients. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
50. Targeting the Hematopoietic Stem Cell Niche in β-Thalassemia and Sickle Cell Disease.
- Author
-
Aprile, Annamaria, Sighinolfi, Silvia, Raggi, Laura, and Ferrari, Giuliana
- Subjects
- *
SICKLE cell anemia , *HEMATOPOIETIC stem cells , *STEM cell niches , *TREATMENT effectiveness , *BONE marrow , *FETAL hemoglobin - Abstract
In the last decade, research on pathophysiology and therapeutic solutions for β-thalassemia (BThal) and sickle cell disease (SCD) has been mostly focused on the primary erythroid defect, thus neglecting the study of hematopoietic stem cells (HSCs) and bone marrow (BM) microenvironment. The quality and engraftment of HSCs depend on the BM microenvironment, influencing the outcome of HSC transplantation (HSCT) both in allogeneic and in autologous gene therapy settings. In BThal and SCD, the consequences of severe anemia alter erythropoiesis and cause chronic stress in different organs, including the BM. Here, we discuss the recent findings that highlighted multiple alterations of the BM niche in BThal and SCD. We point out the importance of improving our understanding of HSC biology, the status of the BM niche, and their functional crosstalk in these disorders towards the novel concept of combined therapies by not only targeting the genetic defect, but also key players of the HSC–niche interaction in order to improve the clinical outcomes of transplantation. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.