Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Nat Immunol ; 23(4): 605-618, 2022 04.
Article in English | MEDLINE | ID: mdl-35352063

ABSTRACT

Autonomic nerves control organ function through the sympathetic and parasympathetic branches, which have opposite effects. In the bone marrow, sympathetic (adrenergic) nerves promote hematopoiesis; however, how parasympathetic (cholinergic) signals modulate hematopoiesis is unclear. Here, we show that B lymphocytes are an important source of acetylcholine, a neurotransmitter of the parasympathetic nervous system, which reduced hematopoiesis. Single-cell RNA sequencing identified nine clusters of cells that expressed the cholinergic α7 nicotinic receptor (Chrna7) in the bone marrow stem cell niche, including endothelial and mesenchymal stromal cells (MSCs). Deletion of B cell-derived acetylcholine resulted in the differential expression of various genes, including Cxcl12 in leptin receptor+ (LepR+) stromal cells. Pharmacologic inhibition of acetylcholine signaling increased the systemic supply of inflammatory myeloid cells in mice and humans with cardiovascular disease.


Subject(s)
Acetylcholine , Hematopoiesis , Animals , B-Lymphocytes , Cholinergic Agents , Hematopoiesis/genetics , Mice , Stem Cell Niche
2.
Cell ; 177(7): 1915-1932.e16, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31130381

ABSTRACT

Stroma is a poorly defined non-parenchymal component of virtually every organ with key roles in organ development, homeostasis, and repair. Studies of the bone marrow stroma have defined individual populations in the stem cell niche regulating hematopoietic regeneration and capable of initiating leukemia. Here, we use single-cell RNA sequencing (scRNA-seq) to define a cellular taxonomy of the mouse bone marrow stroma and its perturbation by malignancy. We identified seventeen stromal subsets expressing distinct hematopoietic regulatory genes spanning new fibroblastic and osteoblastic subpopulations including distinct osteoblast differentiation trajectories. Emerging acute myeloid leukemia impaired mesenchymal osteogenic differentiation and reduced regulatory molecules necessary for normal hematopoiesis. These data suggest that tissue stroma responds to malignant cells by disadvantaging normal parenchymal cells. Our taxonomy of the stromal compartment provides a comprehensive bone marrow cell census and experimental support for cancer cell crosstalk with specific stromal elements to impair normal tissue function and thereby enable emergent cancer.


Subject(s)
Bone Marrow Cells/metabolism , Cell Differentiation , Homeostasis , Leukemia, Myeloid, Acute/metabolism , Osteoblasts/metabolism , Osteogenesis , Tumor Microenvironment , Animals , Bone Marrow Cells/pathology , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Osteoblasts/pathology , Stromal Cells/metabolism , Stromal Cells/pathology
4.
Blood ; 144(1): 21-34, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38579285

ABSTRACT

ABSTRACT: Hematopoietic stem cells (HSCs) are instrumental for organismal survival because they are responsible for lifelong production of mature blood lineages in homeostasis and response to external stress. To fulfill their function, HSCs rely on reciprocal interactions with specialized tissue microenvironments, termed HSC niches. From embryonic development to advanced aging, HSCs transition through several hematopoietic organs in which they are supported by distinct extrinsic cues. Here, we describe recent discoveries on how HSC niches collectively adapt to ensure robust hematopoietic function during biological aging and after exposure to acute stress. We also discuss the latest strategies leveraging niche-derived signals to revert aging-associated phenotypes and enhance hematopoietic recovery after myeloablation.


Subject(s)
Hematopoietic Stem Cells , Stem Cell Niche , Stress, Physiological , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Animals , Humans , Stem Cell Niche/physiology , Adaptation, Physiological , Bone Marrow/pathology , Bone Marrow/metabolism , Bone Marrow/physiology , Aging/physiology , Hematopoiesis/physiology
5.
Nature ; 578(7794): 278-283, 2020 02.
Article in English | MEDLINE | ID: mdl-32025033

ABSTRACT

The biology of haematopoietic stem cells (HSCs) has predominantly been studied under transplantation conditions1,2. It has been particularly challenging to study dynamic HSC behaviour, given that the visualization of HSCs in the native niche in live animals has not, to our knowledge, been achieved. Here we describe a dual genetic strategy in mice that restricts reporter labelling to a subset of the most quiescent long-term HSCs (LT-HSCs) and that is compatible with current intravital imaging approaches in the calvarial bone marrow3-5. We show that this subset of LT-HSCs resides close to both sinusoidal blood vessels and the endosteal surface. By contrast, multipotent progenitor cells (MPPs) show greater variation in distance from the endosteum and are more likely to be associated with transition zone vessels. LT-HSCs are not found in bone marrow niches with the deepest hypoxia and instead are found in hypoxic environments similar to those of MPPs. In vivo time-lapse imaging revealed that LT-HSCs at steady-state show limited motility. Activated LT-HSCs show heterogeneous responses, with some cells becoming highly motile and a fraction of HSCs expanding clonally within spatially restricted domains. These domains have defined characteristics, as HSC expansion is found almost exclusively in a subset of bone marrow cavities with bone-remodelling activity. By contrast, cavities with low bone-resorbing activity do not harbour expanding HSCs. These findings point to previously unknown heterogeneity within the bone marrow microenvironment, imposed by the stages of bone turnover. Our approach enables the direct visualization of HSC behaviours and dissection of heterogeneity in HSC niches.


Subject(s)
Hematopoietic Stem Cells/metabolism , Molecular Imaging , Animals , Bone Remodeling , Cell Movement , Cell Proliferation , Cell Survival , Female , Genes, Reporter , Hypoxia/metabolism , MDS1 and EVI1 Complex Locus Protein/genetics , MDS1 and EVI1 Complex Locus Protein/metabolism , Male , Mice , Oxygen/metabolism , Skull/cytology , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
6.
Nature ; 573(7775): E5, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31515536

ABSTRACT

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

7.
Nature ; 573(7774): 426-429, 2019 09.
Article in English | MEDLINE | ID: mdl-31485073

ABSTRACT

Haematopoietic stem cells self-renew and differentiate into all blood lineages throughout life, and can repair damaged blood systems upon transplantation. Asymmetric cell division has previously been suspected to be a regulator of haematopoietic-stem-cell fate, but its existence has not directly been shown1. In asymmetric cell division, asymmetric fates of future daughter cells are prospectively determined by a mechanism that is linked to mitosis. This can be mediated by asymmetric inheritance of cell-extrinsic niche signals by, for example, orienting the divisional plane, or by the asymmetric inheritance of cell-intrinsic fate determinants. Observations of asymmetric inheritance or of asymmetric daughter-cell fates alone are not sufficient to demonstrate asymmetric cell division2. In both cases, sister-cell fates could be controlled by mechanisms that are independent of division. Here we demonstrate that the cellular degradative machinery-including lysosomes, autophagosomes, mitophagosomes and the protein NUMB-can be asymmetrically inherited into haematopoietic-stem-cell daughter cells. This asymmetric inheritance predicts the asymmetric future metabolic and translational activation and fates of haematopoietic-stem-cell daughter cells and their offspring. Therefore, our studies provide evidence for the existence of asymmetric cell division in haematopoietic stem cells.

8.
Int J Mol Sci ; 24(12)2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37373202

ABSTRACT

Metastasis, particularly brain metastasis, continues to puzzle researchers to this day, and exploring its molecular basis promises to break ground in developing new strategies for combatting this deadly cancer. In recent years, the research focus has shifted toward the earliest steps in the formation of metastasis. In this regard, significant progress has been achieved in understanding how the primary tumor affects distant organ sites before the arrival of tumor cells. The term pre-metastatic niche was introduced for this concept and encompasses all influences on sites of future metastases, ranging from immunological modulation and ECM remodeling to the softening of the blood-brain barrier. The mechanisms governing the spread of metastasis to the brain remain elusive. However, we begin to understand these processes by looking at the earliest steps in the formation of metastasis. This review aims to present recent findings on the brain pre-metastatic niche and to discuss existing and emerging methods to further explore the field. We begin by giving an overview of the pre-metastatic and metastatic niches in general before focusing on their manifestations in the brain. To conclude, we reflect on the methods usually employed in this field of research and discuss novel approaches in imaging and sequencing.


Subject(s)
Brain Neoplasms , Tumor Microenvironment , Humans , Brain , Blood-Brain Barrier , Neoplasm Metastasis
9.
Blood ; 136(20): 2296-2307, 2020 11 12.
Article in English | MEDLINE | ID: mdl-32766876

ABSTRACT

The exact localization of hematopoietic stem cells (HSCs) in their native bone marrow (BM) microenvironment remains controversial, because multiple cell types have been reported to physically associate with HSCs. In this study, we comprehensively quantified HSC localization with up to 4 simultaneous (9 total) BM components in 152 full-bone sections from different bone types and 3 HSC reporter lines. We found adult femoral α-catulin-GFP+ or Mds1GFP/+Flt3Cre HSCs proximal to sinusoids, Cxcl12 stroma, megakaryocytes, and different combinations of those populations, but not proximal to bone, adipocyte, periarteriolar, or Schwann cells. Despite microanatomical differences in femurs and sterna, their adult α-catulin-GFP+ HSCs had similar distributions. Importantly, their microenvironmental localizations were not different from those of random dots, reflecting the relative abundance of imaged BM populations rather than active enrichment. Despite their functional heterogeneity, dormant label-retaining (LR) and non-LR hematopoietic stem and progenitor cells both had indistinguishable localization from α-catulin-GFP+ HSCs. In contrast, cycling juvenile BM HSCs preferentially located close to Cxcl12 stroma and farther from sinusoids/megakaryocytes. We expect our study to help resolve existing confusion regarding the exact localization of different HSC types, their physical association with described BM populations, and their tissue-wide combinations.


Subject(s)
Adult Stem Cells/cytology , Hematopoietic Stem Cells/cytology , Stem Cell Niche , Animals , Mice
10.
Nature ; 535(7611): 299-302, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27411635

ABSTRACT

The mechanisms underlying haematopoietic lineage decisions remain disputed. Lineage-affiliated transcription factors with the capacity for lineage reprogramming, positive auto-regulation and mutual inhibition have been described as being expressed in uncommitted cell populations. This led to the assumption that lineage choice is cell-intrinsically initiated and determined by stochastic switches of randomly fluctuating cross-antagonistic transcription factors. However, this hypothesis was developed on the basis of RNA expression data from snapshot and/or population-averaged analyses. Alternative models of lineage choice therefore cannot be excluded. Here we use novel reporter mouse lines and live imaging for continuous single-cell long-term quantification of the transcription factors GATA1 and PU.1 (also known as SPI1). We analyse individual haematopoietic stem cells throughout differentiation into megakaryocytic-erythroid and granulocytic-monocytic lineages. The observed expression dynamics are incompatible with the assumption that stochastic switching between PU.1 and GATA1 precedes and initiates megakaryocytic-erythroid versus granulocytic-monocytic lineage decision-making. Rather, our findings suggest that these transcription factors are only executing and reinforcing lineage choice once made. These results challenge the current prevailing model of early myeloid lineage choice.


Subject(s)
Cell Differentiation , Cell Lineage , GATA1 Transcription Factor/metabolism , Myeloid Cells/cytology , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Erythrocytes/cytology , Feedback, Physiological , Female , Genes, Reporter , Granulocytes/cytology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Male , Megakaryocytes/cytology , Mice , Models, Biological , Monocytes/cytology , Reproducibility of Results , Single-Cell Analysis , Stochastic Processes
11.
Nat Methods ; 15(1): 39-46, 2018 01.
Article in English | MEDLINE | ID: mdl-29320487

ABSTRACT

Multicolor 3D quantitative imaging of large tissue volumes is necessary to understand tissue development and organization as well as interactions between distinct cell types in situ. However, tissue imaging remains technically challenging, particularly imaging of bone and marrow. Here, we describe a pipeline to reproducibly generate high-dimensional quantitative data from bone and bone marrow that may be extended to any tissue. We generate thick bone sections from adult mouse femurs with preserved tissue microarchitecture and demonstrate eight-color imaging using confocal microscopy without linear unmixing. We introduce XiT, an open-access software for fast and easy data curation, exploration and quantification of large imaging data sets with single-cell resolution. We describe how XiT can be used to correct for potential artifacts in quantitative 3D imaging, and we use the pipeline to measure the spatial relationship between hematopoietic cells, bone matrix and marrow Schwann cells.


Subject(s)
Bone Marrow/ultrastructure , Bone and Bones/ultrastructure , Image Cytometry/methods , Imaging, Three-Dimensional/methods , Microscopy, Confocal/methods , Software , Animals , Male , Mice
12.
Blood ; 133(8): 816-819, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30301719

ABSTRACT

The molecular mechanisms governing the transition from hematopoietic stem cells (HSCs) to lineage-committed progenitors remain poorly understood. Transcription factors (TFs) are powerful cell intrinsic regulators of differentiation and lineage commitment, while cytokine signaling has been shown to instruct the fate of progenitor cells. However, the direct regulation of differentiation-inducing hematopoietic TFs by cell extrinsic signals remains surprisingly difficult to establish. PU.1 is a master regulator of hematopoiesis and promotes myeloid differentiation. Here we report that tumor necrosis factor (TNF) can directly and rapidly upregulate PU.1 protein in HSCs in vitro and in vivo. We demonstrate that in vivo, niche-derived TNF is the principal PU.1 inducing signal in HSCs and is both sufficient and required to relay signals from inflammatory challenges to HSCs.


Subject(s)
Cell Differentiation , Hematopoietic Stem Cells/metabolism , Myelopoiesis , Proto-Oncogene Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Hematopoietic Stem Cells/pathology , Inflammation/metabolism , Inflammation/pathology , Mice , Stem Cell Niche
13.
Proc Natl Acad Sci U S A ; 115(25): E5688-E5695, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29866839

ABSTRACT

In adults, human hematopoietic stem and progenitor cells (HSPCs) reside in the bone marrow (BM) microenvironment. Our understanding of human hematopoiesis and the associated niche biology remains limited, due to human material accessibility and limits of existing in vitro culture models. The establishment of an in vitro BM system would offer an experimentally accessible and tunable platform to study human hematopoiesis. Here, we develop a 3D engineered human BM analog by recapitulating some of the hematopoietic niche elements. This includes a bone-like scaffold, functionalized by human stromal and osteoblastic cells and by the extracellular matrix they deposited during perfusion culture in bioreactors. The resulting tissue exhibited compositional and structural features of human BM while supporting the maintenance of HSPCs. This was associated with a compartmentalization of phenotypes in the bioreactor system, where committed blood cells are released into the liquid phase and HSPCs preferentially reside within the engineered BM tissue, establishing physical interactions with the stromal compartment. Finally, we demonstrate the possibility to perturb HSPCs' behavior within our 3D niches by molecular customization or injury simulation. The developed system enables the design of advanced, tunable in vitro BM proxies for the study of human hematopoiesis.


Subject(s)
Bone Marrow Cells/cytology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Stem Cell Niche/physiology , Stem Cells/cytology , Biomimetics/methods , Bioreactors , Bone Marrow/physiology , Cell Culture Techniques/methods , Extracellular Matrix/physiology , Humans , Tissue Engineering/methods
14.
Curr Opin Oncol ; 32(2): 154-161, 2020 03.
Article in English | MEDLINE | ID: mdl-32022758

ABSTRACT

PURPOSE OF REVIEW: Hematopoietic stem cells (HSCs) reside in specialized anatomical microenvironments within the bone marrow space, termed HSC niches. Different bone marrow imaging modalities have been utilized to visualize HSCs in situ, and unravel the cellular identity of bone marrow cell types located in their immediate proximity. However, despite extensive research, the exact identity of bone marrow populations that physically associate with HSCs remains controversial. RECENT FINDINGS: Recent advances in volumetric imaging enable precise identification of bone marrow populations and their spatial distribution both at tissue-wide scale and single-cell resolution. In addition, single-cell RNA sequencing and mass-cytometry-based approaches dissect the complexity of the bone marrow microenvironment with unprecedented resolution. Here, we review current concepts regarding bone marrow populations that physically associate with HSCs and recent efforts to localize HSCs and their niche populations. SUMMARY: Defining the bone marrow cell types in the immediate proximity of HSCs in homeostasis and stress is key to determine the cellular and molecular cues driving HSC maintenance and regeneration.


Subject(s)
Bone Marrow Cells/cytology , Hematopoietic Stem Cells/cytology , Animals , Cellular Microenvironment/physiology , Humans , Stem Cell Niche
15.
Nat Methods ; 14(4): 403-406, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28218899

ABSTRACT

Differentiation alters molecular properties of stem and progenitor cells, leading to changes in their shape and movement characteristics. We present a deep neural network that prospectively predicts lineage choice in differentiating primary hematopoietic progenitors using image patches from brightfield microscopy and cellular movement. Surprisingly, lineage choice can be detected up to three generations before conventional molecular markers are observable. Our approach allows identification of cells with differentially expressed lineage-specifying genes without molecular labeling.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Image Processing, Computer-Assisted/methods , Neural Networks, Computer , Time-Lapse Imaging/methods , Animals , Area Under Curve , Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Gene Knock-In Techniques , Machine Learning , Male , Mice, Mutant Strains , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
16.
Blood ; 129(12): 1691-1701, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28159742

ABSTRACT

Controlled regulation of lineage decisions is imperative for hematopoiesis. Yet, the molecular mechanisms underlying hematopoietic lineage choices are poorly defined. Colony-stimulating factor 1 (CSF-1), the cytokine acting as the principal regulator of monocyte/macrophage (M) development, has been shown to be able to instruct the lineage choice of uncommitted granulocyte M (GM) progenitors toward an M fate. However, the intracellular signaling pathways involved are unknown. CSF-1 activates a multitude of signaling pathways resulting in a pleiotropic cellular response. The precise role of individual pathways within this complex and redundant signaling network is dependent on cellular context, and is not well understood. Here, we address which CSF-1-activated pathways are involved in transmitting the lineage-instructive signal in primary bone marrow-derived GM progenitors. Although its loss is compensated for by alternative signaling activation mechanisms, Src family kinase (SFK) signaling is sufficient to transmit the CSF-1 lineage instructive signal. Moreover, c-Src activity is sufficient to drive M fate, even in nonmyeloid cells.


Subject(s)
Cell Lineage , Macrophage Colony-Stimulating Factor/physiology , Monocytes/cytology , Signal Transduction , src-Family Kinases/metabolism , Animals , Cells, Cultured , Granulocyte Precursor Cells/cytology , Hematopoiesis , Mice
17.
Anal Chem ; 90(18): 10695-10700, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30059208

ABSTRACT

Dynamic environments determine cell fate decisions and function. Understanding the relationship between extrinsic signals on cellular responses and cell fate requires the ability to dynamically change environmental inputs in vitro, while continuously observing individual cells over extended periods of time. This is challenging for nonadherent cells, such as hematopoietic stem and progenitor cells, because media flow displaces and disturbs such cells, preventing culture and tracking of single cells. Here, we present a programmable microfluidic system designed for the long-term culture and time-lapse imaging of nonadherent cells in dynamically changing cell culture conditions without losing track of individual cells. The dynamic, valve-controlled design permits targeted seeding of cells in up to 48 independently controlled culture chambers, each providing sufficient space for long-term cell colony expansion. Diffusion-based media exchange occurs rapidly and minimizes displacement of cells and eliminates shear stress. The chip was successfully tested with long-term culture and tracking of primary hematopoietic stem and progenitor cells, and murine embryonic stem cells. This system will have important applications to analyze dynamic signaling inputs controlling fate choices.


Subject(s)
Cell Tracking/methods , Hematopoietic Stem Cells/cytology , Lab-On-A-Chip Devices , Mouse Embryonic Stem Cells/cytology , Single-Cell Analysis/methods , Animals , Cell Adhesion , Cells, Cultured , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/metabolism , Proof of Concept Study , Reproducibility of Results , Time-Lapse Imaging
18.
Blood ; 128(9): 1181-92, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27365423

ABSTRACT

The maintenance of hematopoietic stem cells (HSCs) during ex vivo culture is an important prerequisite for their therapeutic manipulation. However, despite intense research, culture conditions for robust maintenance of HSCs are still missing. Cultured HSCs are quickly lost, preventing their improved analysis and manipulation. Identification of novel factors supporting HSC ex vivo maintenance is therefore necessary. Coculture with the AFT024 stroma cell line is capable of maintaining HSCs ex vivo long-term, but the responsible molecular players remain unknown. Here, we use continuous long-term single-cell observation to identify the HSC behavioral signature under supportive or nonsupportive stroma cocultures. We report early HSC survival as a major characteristic of HSC-maintaining conditions. Behavioral screening after manipulation of candidate molecules revealed that the extracellular matrix protein dermatopontin (Dpt) is involved in HSC maintenance. DPT knockdown in supportive stroma impaired HSC survival, whereas ectopic expression of the Dpt gene or protein in nonsupportive conditions restored HSC survival. Supplementing defined stroma- and serum-free culture conditions with recombinant DPT protein improved HSC clonogenicity. These findings illustrate a previously uncharacterized role of Dpt in maintaining HSCs ex vivo.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Extracellular Matrix Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Animals , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Chondroitin Sulfate Proteoglycans/genetics , Chondroitin Sulfate Proteoglycans/pharmacology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/pharmacology , Hematopoietic Stem Cells/cytology , Male , Mice , Mice, Transgenic , Stromal Cells/cytology , Stromal Cells/metabolism , Time Factors
20.
Electromagn Biol Med ; 33(3): 165-89, 2014 Sep.
Article in English | MEDLINE | ID: mdl-23915130

ABSTRACT

The model biological organisms Drosophila melanogaster and Drosophila virilis have been utilized to assess effects on apoptotic cell death of follicles during oogenesis and reproductive capacity (fecundity) decline. A total of 280 different experiments were performed using newly emerged flies exposed for short time daily for 3-7 d to various EMF sources including: GSM 900/1800 MHz mobile phone, 1880-1900 MHz DECT wireless base, DECT wireless handset, mobile phone-DECT handset combination, 2.44 GHz wireless network (Wi-Fi), 2.44 GHz blue tooth, 92.8 MHz FM generator, 27.15 MHz baby monitor, 900 MHz CW RF generator and microwave oven's 2.44 GHz RF and magnetic field components. Mobile phone was used as a reference exposure system for evaluating factors considered very important in dosimetry extending our published work with D. melanogaster to the insect D. virilis. Distance from the emitting source, the exposure duration and the repeatability were examined. All EMF sources used created statistically significant effects regarding fecundity and cell death-apoptosis induction, even at very low intensity levels (0.3 V/m blue tooth radiation), well below ICNIRP's guidelines, suggesting that Drosophila oogenesis system is suitable to be used as a biomarker for exploring potential EMF bioactivity. Also, there is no linear cumulative effect when increasing the duration of exposure or using one EMF source after the other (i.e. mobile phone and DECT handset) at the specific conditions used. The role of the average versus the peak E-field values as measured by spectrum analyzers on the final effects is discussed.


Subject(s)
Drosophila melanogaster/physiology , Drosophila melanogaster/radiation effects , Electromagnetic Fields/adverse effects , Oogenesis/radiation effects , Animals , Apoptosis/radiation effects , Biomarkers , Cell Phone/instrumentation , Drosophila melanogaster/cytology , Environmental Exposure/adverse effects , Female , Microwaves , Ovarian Follicle/cytology , Ovarian Follicle/physiology , Ovarian Follicle/radiation effects , Pupa/physiology , Pupa/radiation effects , Radiation Protection/instrumentation , Radio/instrumentation , Wireless Technology/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL