Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Lett ; 488: 9-17, 2020 09 28.
Article in English | MEDLINE | ID: mdl-32479768

ABSTRACT

Breast cancer (BC) remains a clinical challenge despite improved treatments and public awareness to ensure early diagnosis. A major issue is the ability of BC cells (BCCs) to survive as dormant cancer cells in the bone marrow (BM), resulting in the cancer surviving for decades with the potential to resurge as metastatic cancer. The experimental evidence indicates similarity between dormant BCCs and other stem cells, resulting in the preponderance of data to show dormant BCCs being cancer stem cells (CSCs). The BM niche and their secretome support BCC dormancy. Lacking in the literature is a comprehensive research to describe how the hypoxic environment within the BM may influence the behavior of BCCs. This information is relevant to understand the prognosis of BC in young and aged individuals whose oxygen levels differ in BM. This review discusses the changing information on vascularity in different regions of the BM and the impact on endogenous hematopoietic stem cells (HSCs). This review highlights the necessary information to provide insights on vascularity of different BM regions on the behavior of BCCs, in particular a dormant phase. For instance, how the transcription factor HIF1-α (hypoxia-inducible factor 1 alpha), functioning as first responder under hypoxic conditions, affects the expression of specific gene networks involved in energy metabolism, cell survival, tumor invasion and angiogenesis. This enables cell fate transition and facilitates tumor heterogeneity, which in turn favors tumor progression and resistance to anticancer treatments Thus, HIF1-α could be a potential target for cancer treatment. This review describes epigenetic mechanisms involved in hypoxic responses during cancer dormancy in the bone marrow. The varied hypoxic environment in the BM is relevant to understand the complex process of the aging bone marrow for insights on breast cancer outcome between the young and aged.


Subject(s)
Bone Marrow Neoplasms/secondary , Breast Neoplasms/pathology , Cell Hypoxia/physiology , Neoplastic Stem Cells/pathology , Tumor Microenvironment/physiology , Animals , Bone Marrow/pathology , Female , Humans
2.
J Immunol ; 204(4): 879-891, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31924647

ABSTRACT

Hematopoiesis is tightly regulated by the bone marrow (BM) niche. The niche is robust, allowing for the return of hematopoietic homeostasis after insults such as infection. Hematopoiesis is partly regulated by soluble factors, such as neuropeptides, substance P (SP), and neurokinin A (NK-A), which mediate hematopoietic stimulation and inhibition, respectively. SP and NK-A are derived from the Tac1 gene that is alternately spliced into four variants. The hematopoietic effects of SP and NK-A are mostly mediated via BM stroma. Array analyses with 2400 genes indicated distinct changes in SP-stimulated BM stroma. Computational analyses indicated networks of genes with hematopoietic regulation. Included among these networks is the high-mobility group box 1 gene (HMGB1), a nonhistone chromatin-associated protein. Validation studies indicated that NK-A could reverse SP-mediated HMGB1 decrease. Long-term culture-initiating cell assay, with or without NK-A receptor antagonist (NK2), showed a suppressive effect of HMGB1 on hematopoietic progenitors and increase in long-term culture-initiating cell assay cells (primitive hematopoietic cells). These effects occurred partly through NK-A. NSG mice with human hematopoietic system injected with the HMGB1 antagonist glycyrrhizin verified the in vitro effects of HMGB1. Although the effects on myeloid lineage were suppressed, the results suggested a more complex effect on the lymphoid lineage. Clonogenic assay for CFU- granulocyte-monocyte suggested that HMGB1 may be required to prevent hematopoietic stem cell exhaustion to ensure immune homeostasis. In summary, this study showed how HMGB1 is linked to SP and NK-A to protect the most primitive hematopoietic cell and also to maintain immune/hematopoietic homeostasis.


Subject(s)
HMGB1 Protein/metabolism , Hematopoiesis/genetics , Neuroimmunomodulation/genetics , Neurokinin A/metabolism , Substance P/metabolism , Adolescent , Adult , Alternative Splicing , Animals , Benzamides/pharmacology , Biopsy , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow Transplantation , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/immunology , HEK293 Cells , Hematopoiesis/immunology , Hematopoietic Stem Cells/metabolism , Humans , Mice , Neuroimmunomodulation/immunology , Neurokinin A/antagonists & inhibitors , Oligonucleotide Array Sequence Analysis , Piperidines/pharmacology , Primary Cell Culture , Tachykinins/genetics , Transplantation Chimera , Young Adult
3.
Adv Exp Med Biol ; 1201: 93-108, 2019.
Article in English | MEDLINE | ID: mdl-31898783

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent cells that can self-renew and differentiate into cells of all germ layers. MSCs can be easily attracted to the site of tissue insult with high levels of inflammatory mediators. The general ability of MSCs to migrate at the sites of tissue injury suggested an innate ability for these cells to be involved in baseline tissue repair. The bone marrow is one of the primary sources of MSCs, though they can be ubiquitous. An attractive property of MSCs for clinical application is their ability to cross allogeneic barrier. However, alone, MSCs are not immune suppressive cells. Rather, they can be licensed by the tissue microenvironment to become immune suppressor cells. Immune suppressor functions of MSCs include those that blunt cytotoxicity of natural killer cells, suppression of T-cell proliferation, and "veto" function. MSCs, as third-party cells, suppress the immune response that generally recapitulates graft-versus-host disease (GvHD) responses. Based on the plastic functions of MSCs, these cells have dominated the field of cell-based therapies, such as anti-inflammatory and drug delivery. Here, we focus on the potential use of MSC for immunological disorders such as Crohn's disease and GvHD.


Subject(s)
Immune System Diseases/immunology , Immune System Diseases/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Crohn Disease/immunology , Crohn Disease/therapy , Graft vs Host Disease/immunology , Graft vs Host Disease/therapy , Humans , Immune System Diseases/pathology , Killer Cells, Natural/immunology , Mesenchymal Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...