Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Angiogenesis ; 26(1): 129-166, 2023 02.
Article in English | MEDLINE | ID: mdl-36183032

ABSTRACT

Cancer cells are embedded within the tissue and interact dynamically with its components during cancer progression. Understanding the contribution of cellular components within the tumor microenvironment is crucial for the success of therapeutic applications. Here, we reveal the presence of perivascular GFAP+/Plp1+ cells within the tumor microenvironment. Using in vivo inducible Cre/loxP mediated systems, we demonstrated that these cells derive from tissue-resident Schwann cells. Genetic ablation of endogenous Schwann cells slowed down tumor growth and angiogenesis. Schwann cell-specific depletion also induced a boost in the immune surveillance by increasing tumor-infiltrating anti-tumor lymphocytes, while reducing immune-suppressor cells. In humans, a retrospective in silico analysis of tumor biopsies revealed that increased expression of Schwann cell-related genes within melanoma was associated with improved survival. Collectively, our study suggests that Schwann cells regulate tumor progression, indicating that manipulation of Schwann cells may provide a valuable tool to improve cancer patients' outcomes.


Subject(s)
Neoplasms , Neuroglia , Humans , Retrospective Studies , Neuroglia/metabolism , Schwann Cells/metabolism , Schwann Cells/pathology , Pericytes , Tumor Microenvironment/physiology , Neoplasms/pathology
2.
Acta Neuropathol Commun ; 9(1): 183, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34784974

ABSTRACT

Sensory neurons have recently emerged as components of the tumor microenvironment. Nevertheless, whether sensory neuronal activity is important for tumor progression remains unknown. Here we used Designer Receptors Exclusively Activated by a Designer Drug (DREADD) technology to inhibit or activate sensory neurons' firing within the melanoma tumor. Melanoma growth and angiogenesis were accelerated following inhibition of sensory neurons' activity and were reduced following overstimulation of these neurons. Sensory neuron-specific overactivation also induced a boost in the immune surveillance by increasing tumor-infiltrating anti-tumor lymphocytes, while reducing immune-suppressor cells. In humans, a retrospective in silico analysis of melanoma biopsies revealed that increased expression of sensory neurons-related genes within melanoma was associated with improved survival. These findings suggest that sensory innervations regulate melanoma progression, indicating that manipulation of sensory neurons' activity may provide a valuable tool to improve melanoma patients' outcomes.


Subject(s)
Melanoma/genetics , Melanoma/pathology , Sensory Receptor Cells/pathology , Animals , Behavior, Animal/drug effects , Biopsy , Cell Line, Tumor , Computer Simulation , Disease Progression , Humans , Immunologic Surveillance , Lymphocytes/pathology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Transgenic , NAV1.8 Voltage-Gated Sodium Channel/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Sensory Receptor Cells/metabolism , Suppressor Factors, Immunologic , Tumor Microenvironment
3.
mBio ; 12(5): e0130721, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34607465

ABSTRACT

Toxoplasmosis affects one-third of the human population worldwide. Humans are accidental hosts and are infected after consumption of undercooked meat and water contaminated with Toxoplasma gondii cysts and oocysts, respectively. Neutrophils have been shown to participate in the control of T. gondii infection in mice through a variety of effector mechanisms, such as reactive oxygen species (ROS) and neutrophil extracellular trap (NET) formation. However, few studies have demonstrated the role of neutrophils in individuals naturally infected with T. gondii. In the current study, we evaluated the activation status of neutrophils in individuals with acute or chronic toxoplasmosis and determined the role of T. gondii-induced NET formation in the amplification of the innate and adaptive immune responses. We observed that neutrophils are highly activated during acute infection through increased expression of CD66b. Moreover, neutrophils from healthy donors (HDs) cocultured with tachyzoites produced ROS and formed NETs, with the latter being dependent on glycolysis, succinate dehydrogenase, gasdermin D, and neutrophil elastase. Furthermore, we observed elevated levels of the chemokines (CXC motif) CXCL8 and (CC motif) CCL4 ligands in plasma from patients with acute toxoplasmosis and production by neutrophils from HDs exposed to T. gondii. Finally, we showed that T. gondii-induced NETs activate neutrophils and promote the recruitment of autologous CD4+ T cells and the production of interferon gamma (IFN-γ), tumor necrosis factor (TNF), interleukin 6 (IL-6), IL-17, and IL-10 by peripheral blood mononuclear cells. In conclusion, we demonstrated that T. gondii activates neutrophils and promotes the release of NETs, which amplify human innate and adaptive immune responses. IMPORTANCE Approximately one-third of the human population is estimated to be chronically infected with the obligate intracellular parasite Toxoplasma gondii. Humans are accidental hosts that are infected with T. gondii after consumption of undercooked meat or contaminated water. Neutrophils have been shown to control T. gondii growth by different mechanisms, including neutrophil extracellular traps (NETs). In the current study, we observed that neutrophils are highly activated during acute toxoplasmosis. We also determined that T. gondii-induced NETs are dependent on the energetic profile of neutrophils as well as the production of ROS and gasdermin D (GSDMD) cleavage. In addition, we showed that T. gondii-induced NETs activate neutrophils, promote the recruitment of autologous CD4+ T cells, and induce the production of cytokines by peripheral blood mononuclear cells, amplifying the innate and adaptive immune responses.


Subject(s)
Adaptive Immunity , Extracellular Traps/immunology , Immunity, Innate , Neutrophils/immunology , Toxoplasma/immunology , Adult , Antigens, CD/genetics , Antigens, CD/immunology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Chemokines/immunology , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Humans , Interleukins/classification , Interleukins/immunology , Leukocytes, Mononuclear/immunology , Male , Neutrophils/parasitology , Young Adult
4.
Crit Rev Oncol Hematol ; 163: 103368, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34051302

ABSTRACT

Hematopoietic stem cells are the most illustrious inhabitants of the bone marrow. Direct visualization of endogenous hematopoietic stem cells in this niche is essential to study their functions. Until recently this was not possible in live animals. Recent studies, using state-of-the-art technologies, including sophisticated in vivo inducible genetic approaches in combination with two-photon laser scanning microscopy, allow the follow-up of endogenous hematopoietic stem cells' behavior in their habitat. Strikingly, the new findings reveal that quiescent hematopoietic stem cells are more mobile than previously thought, and link their retained steady state within the niche to a mobile behavior. The arising knowledge from this research will be critical for the therapy of several hematological diseases. Here, we review recent progress in our understanding of hematopoietic stem cell biology in their niches.


Subject(s)
Bone Marrow , Stem Cell Niche , Animals , Bone Marrow Cells , Cell Division , Hematopoietic Stem Cells , Humans
5.
Histochem Cell Biol ; 156(2): 165-182, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34003355

ABSTRACT

Diagnosis and prognosis of breast cancer is based on disease staging identified through histopathological and molecular biology techniques. Animal models are used to gain mechanistic insights into the development of breast cancer. C(3)1-TAg is a genetically engineered mouse model that develops mammary cancer. However, carcinogenesis caused by this transgene was characterized in the Friend Virus B (FVB) background. As most genetic studies are done in mice with C57BL/6 J background, we aimed to define the histological alterations in C3(1)-TAg C57BL/6 J animals. Our results showed that C3(1)-TAg animals with C57BL/6 J background develop solid-basaloid adenoid cystic carcinomas with increased fibrosis, decreased area of adipocytes, and a high proliferative index, which are triple-negative for progesterone, estrogen, and human epidermal growth factor receptor 2 (HER2) receptors. Our results also revealed that tumor development is slower in the C57BL/6 J background when compared with the FVB strain, providing a better model to study the different stages in breast cancer progression.


Subject(s)
Antigens, Viral, Tumor/genetics , Breast Neoplasms/genetics , Carcinoma, Adenoid Cystic/genetics , Models, Genetic , Animals , Antigens, Viral, Tumor/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Carcinoma, Adenoid Cystic/immunology , Carcinoma, Adenoid Cystic/pathology , Female , Friend murine leukemia virus/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
6.
Stem Cell Rev Rep ; 17(5): 1874-1888, 2021 10.
Article in English | MEDLINE | ID: mdl-34003465

ABSTRACT

Multiple infectious diseases lead to impaired lung function. Revealing the cellular mechanisms involved in this impairment is crucial for the understanding of how the lungs shift from a physiologic to a pathologic state in each specific condition. In this context, we explored the pathogenesis of Paracoccidioidomycosis, which affects pulmonary functioning. The presence of cells expressing Nestin-GFP has been reported in different tissues, and their roles as tissue-specific progenitors have been stablished in particular organs. Here, we explored how Nestin-GFP+ cells are affected after lung infection by Paracoccidioides brasiliensis, a model of lung granulomatous inflammation with fibrotic outcome. We used Nestin-GFP transgenic mice, parabiosis surgery, confocal microscopy and flow cytometry to investigate the participation of Nestin-GFP+ cells in Paracoccidioides brasiliensis pathogenesis. We revealed that these cells increase in the lungs post-Paracoccidioides brasiliensis infection, accumulating around granulomas. This increase was due mainly to Nestin-GPF+ cells derived from the blood circulation, not associated to blood vessels, that co-express markers suggestive of hematopoietic cells (Sca-1, CD45 and CXCR4). Therefore, our findings suggest that circulating Nestin-GFP+ cells participate in the Paracoccidioides brasiliensis pathogenesis in the lungs.


Subject(s)
Lung , Animals , Mice , Nestin/genetics , Paracoccidioides/genetics
7.
Stem Cells Transl Med ; 10(3): 346-356, 2021 03.
Article in English | MEDLINE | ID: mdl-33112056

ABSTRACT

Niches are specialized tissue microenvironments that control stem cells functioning. The bone marrow mesenchymal stem cell niche defines a location within the marrow in which mesenchymal stem cells are retained and produce new cells throughout life. Deciphering the signaling mechanisms by which the niche regulates stem cell fate will facilitate the use of these cells for therapy. Recent studies, by using state-of-the-art methodologies, including sophisticated in vivo inducible genetic techniques, such as lineage-tracing Cre/loxP mediated systems, in combination with pharmacological inhibition, provide evidence that sensory neuron is an important component of the bone marrow mesenchymal stem cell niche. Strikingly, knockout of a specific receptor in sensory neurons blocked stem cell function in the bone marrow. The knowledge arising from these discoveries will be crucial for stem cell manipulation in the future. Here, we review recent progress in our understanding of sensory nerves biology in the stem cell niche.


Subject(s)
Mesenchymal Stem Cells , Sensory Receptor Cells , Stem Cell Niche , Bone Marrow , Cell Differentiation , Stem Cells
8.
J Cell Mol Med ; 24(17): 9574-9589, 2020 09.
Article in English | MEDLINE | ID: mdl-32691511

ABSTRACT

The tumour mass is composed not only of heterogeneous neoplastic cells, but also a variety of other components that may affect cancer cells behaviour. The lack of detailed knowledge about all the constituents of the tumour microenvironment restricts the design of effective treatments. Nerves have been reported to contribute to the growth and maintenance of numerous tissues. The effects of sensory innervations on tumour growth remain unclear. Here, by using state-of-the-art techniques, including Cre/loxP technologies, confocal microscopy, in vivo-tracing and chemical denervation, we revealed the presence of sensory nerves infiltrating within the melanoma microenvironment, and affecting cancer progression. Strikingly, melanoma growth in vivo was accelerated following genetic ablation or chemical denervation of sensory nerves. In humans, a retrospective analysis of melanoma patients revealed that increased expression of genes related to sensory nerves in tumours was associated with better clinical outcomes. These findings suggest that sensory innervations counteract melanoma progression. The emerging knowledge from this research provides a novel target in the tumour microenvironment for therapeutic benefit in cancer patients.


Subject(s)
Melanoma/pathology , Sensory Receptor Cells/pathology , Skin Neoplasms/pathology , Animals , Cell Communication/physiology , Cell Line, Tumor , Disease Progression , Humans , Mice , Mice, Inbred C57BL , Retrospective Studies , Tumor Microenvironment
10.
Nat Chem Biol ; 16(8): 920-929, 2020 08.
Article in English | MEDLINE | ID: mdl-32451508

ABSTRACT

The fundamental and assorted roles of ß-1,3-glucans in nature are underpinned on diverse chemistry and molecular structures, demanding sophisticated and intricate enzymatic systems for their processing. In this work, the selectivity and modes of action of a glycoside hydrolase family active on ß-1,3-glucans were systematically investigated combining sequence similarity network, phylogeny, X-ray crystallography, enzyme kinetics, mutagenesis and molecular dynamics. This family exhibits a minimalist and versatile (α/ß)-barrel scaffold, which can harbor distinguishing exo or endo modes of action, including an ancillary-binding site for the anchoring of triple-helical ß-1,3-glucans. The substrate binding occurs via a hydrophobic knuckle complementary to the canonical curved conformation of ß-1,3-glucans or through a substrate conformational change imposed by the active-site topology of some fungal enzymes. Together, these findings expand our understanding of the enzymatic arsenal of bacteria and fungi for the breakdown and modification of ß-1,3-glucans, which can be exploited for biotechnological applications.


Subject(s)
Glucan 1,3-beta-Glucosidase/chemistry , Glycoside Hydrolases/chemistry , beta-Glucans/chemistry , Amino Acid Sequence/genetics , Binding Sites/physiology , Catalytic Domain/physiology , Crystallography, X-Ray/methods , Glucan 1,3-beta-Glucosidase/metabolism , Glucans/chemistry , Glycosides/chemistry , Models, Molecular , Substrate Specificity/physiology
11.
Nat Med ; 24(9): 1330-1336, 2018 09.
Article in English | MEDLINE | ID: mdl-30038217

ABSTRACT

Plasmodium vivax causes approximately 100 million clinical malaria cases yearly1,2. The basis of protective immunity is poorly understood and thought to be mediated by antibodies3,4. Cytotoxic CD8+ T cells protect against other intracellular parasites by detecting parasite peptides presented by human leukocyte antigen class I on host cells. Cytotoxic CD8+ T cells kill parasite-infected mammalian cells and intracellular parasites by releasing their cytotoxic granules5,6. Perforin delivers the antimicrobial peptide granulysin and death-inducing granzymes into the host cell, and granulysin then delivers granzymes into the parasite. Cytotoxic CD8+ T cells were thought to have no role against Plasmodium spp. blood stages because red blood cells generally do not express human leukocyte antigen class I7. However, P. vivax infects reticulocytes that retain the protein translation machinery. Here we show that P. vivax-infected reticulocytes express human leukocyte antigen class I. Infected patient circulating CD8+ T cells highly express cytotoxic proteins and recognize and form immunological synapses with P. vivax-infected reticulocytes in a human leukocyte antigen-dependent manner, releasing their cytotoxic granules to kill both host cell and intracellular parasite, preventing reinvasion. P. vivax-infected reticulocytes and parasite killing is perforin independent, but depends on granulysin, which generally efficiently forms pores only in microbial membranes8. We find that P. vivax depletes cholesterol from the P. vivax-infected reticulocyte cell membrane, rendering it granulysin-susceptible. This unexpected T cell defense might be mobilized to improve P. vivax vaccine efficacy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Plasmodium vivax/physiology , Reticulocytes/parasitology , Antigens, Differentiation, T-Lymphocyte/metabolism , Female , HLA Antigens/metabolism , Humans , Lymphocyte Activation/immunology , Lymphocyte Count , Malaria/blood , Male , Reticulocytes/ultrastructure
12.
J Infect Dis ; 218(8): 1314-1323, 2018 09 08.
Article in English | MEDLINE | ID: mdl-29800313

ABSTRACT

The balance between pro- and antiinflammatory mechanisms is essential to limit immune-mediated pathology, and CD4+ forkhead box P3 (Foxp3+) regulatory T cells (Treg) play an important role in this process. The expression of inhibitory receptors regulates cytokine production by Plasmodium vivax-specific T cells. Our goal was to assess the induction of programmed death-1 (PD-1) and cytotoxic T-lymphocyte antigen (CTLA-4) on Treg during malaria and to evaluate their function. We found that P. vivax infection triggered an increase in circulating Treg and their expression of CTLA-4 and PD-1. Functional analysis demonstrated that Treg from malaria patients had impaired suppressive ability and PD-1+Treg displayed lower levels of Foxp3 and Helios, but had higher frequencies of T-box transcription factor+ and interferon-gamma+ cells than PD-1-Treg. Thus malaria infection alters the function of circulating Treg by triggering increased expression of PD-1 on Treg that is associated with decreased regulatory function and increased proinflammatory characteristics.


Subject(s)
Malaria, Vivax/immunology , Malaria, Vivax/parasitology , T-Lymphocytes, Regulatory/physiology , Adult , Cell Proliferation , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Regulation/immunology , Humans , Immunophenotyping , Male , Middle Aged , Plasmodium vivax , Reticulocytes/parasitology , Reticulocytes/physiology , Young Adult
13.
J Infect Dis ; 212(12): 1999-2010, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26019284

ABSTRACT

The function and regulation of the immune response triggered during malaria is complex and poorly understood, and there is a particular paucity of studies conducted in humans infected with Plasmodium vivax. While it has been proposed that T-cell-effector responses are crucial for protection against blood-stage malaria in mice, the mechanisms behind this in humans remain poorly understood. Experimental models of malaria have shown that the regulatory molecules, cytotoxic T-lymphocyte attenuator-4 (CTLA-4), lymphocyte activation gene-3 (LAG-3), and programmed death-1 (PD-1) are involved in the functional impairment of T cells during infection. Our goal was to define the role of these molecules during P. vivax malaria. We demonstrate that infection triggers the expression of regulatory molecules on T cells. The pattern of expression differs in CD4(+) and CD8(+) T cells. Higher frequencies of CD4(+) express more than 1 regulatory molecule compared to CD8(+) T cells. Moreover, lower proportions of CD4(+) T cells coexpress regulatory molecules, but are still able to proliferate. Importantly, simultaneously blockade of the CLTA-4, PD-1, and T-cell immunoglobulin and mucin-3 signaling restores the cytokine production by antigen-specific cells. These data support the hypothesis that upregulation of inhibitory receptors on T cells during P. vivax malaria impairs parasite-specific T-cell effector function.


Subject(s)
Cytokines/antagonists & inhibitors , Host-Pathogen Interactions , Immune Tolerance , Malaria, Vivax/immunology , Plasmodium vivax/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Adult , Female , Humans , Malaria, Vivax/parasitology , Male , Middle Aged , Young Adult
14.
PLoS Pathog ; 10(9): e1004393, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25233271

ABSTRACT

Infection with Plasmodium vivax results in strong activation of monocytes, which are important components of both the systemic inflammatory response and parasite control. The overall goal of this study was to define the role of monocytes during P. vivax malaria. Here, we demonstrate that P. vivax-infected patients display significant increase in circulating monocytes, which were defined as CD14(+)CD16- (classical), CD14(+)CD16(+) (inflammatory), and CD14loCD16(+) (patrolling) cells. While the classical and inflammatory monocytes were found to be the primary source of pro-inflammatory cytokines, the CD16(+) cells, in particular the CD14(+)CD16(+) monocytes, expressed the highest levels of activation markers, which included chemokine receptors and adhesion molecules. Morphologically, CD14(+) were distinguished from CD14lo monocytes by displaying larger and more active mitochondria. CD14(+)CD16(+) monocytes were more efficient in phagocytizing P. vivax-infected reticulocytes, which induced them to produce high levels of intracellular TNF-α and reactive oxygen species. Importantly, antibodies specific for ICAM-1, PECAM-1 or LFA-1 efficiently blocked the phagocytosis of infected reticulocytes by monocytes. Hence, our results provide key information on the mechanism by which CD14(+)CD16(+) cells control parasite burden, supporting the hypothesis that they play a role in resistance to P. vivax infection.


Subject(s)
Erythrocytes/immunology , Inflammation/immunology , Lipopolysaccharide Receptors/immunology , Malaria, Vivax/immunology , Mitochondria/immunology , Monocytes/immunology , Plasmodium vivax/immunology , Receptors, IgG/immunology , Acute Disease , Adolescent , Adult , Aged , Female , Flow Cytometry , Humans , Immunophenotyping , Malaria, Vivax/metabolism , Malaria, Vivax/parasitology , Male , Middle Aged , Mitochondria/metabolism , Mitochondria/pathology , Monocytes/metabolism , Monocytes/parasitology , Phagocytosis , Reactive Oxygen Species/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...