Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
J Histochem Cytochem ; 70(1): 53-81, 2022 01.
Article in English | MEDLINE | ID: mdl-34751050

ABSTRACT

Immune checkpoint inhibitors have become the mainstay of treatment for hepatocellular carcinoma (HCC). However, they are ineffective in some cases. Previous studies have reported that genetic alterations in oncogenic pathways such as Wnt/ß-catenin are the important triggers in HCC for primary refractoriness. T-cell exhaustion has been reported in various tumors and is likely to play a prominent role in the emergence of HCC due to chronic inflammation and cirrhosis-associated immune dysfunction. Immunosuppressive cells including regulatory T-cells and tumor-associated macrophages infiltrating the tumor are associated with hyperprogressive disease in the early stages of immune checkpoint inhibitor treatment. In addition, stellate cells and tumor-associated fibroblasts create an abundant desmoplastic environment by producing extracellular matrix. This strongly contributes to epithelial to mesenchymal transition via signaling activities including transforming growth factor beta, Wnt/ß-catenin, and Hippo pathway. The abundant desmoplastic environment has been demonstrated in pancreatic ductal adenocarcinoma and cholangiocarcinoma to suppress cytotoxic T-cell infiltration, PD-L1 expression, and neoantigen expression, resulting in a highly immunosuppressive niche. It is possible that a similar immunosuppressive environment is created in HCC with advanced fibrosis in the background liver. Although sufficient understanding is required for the establishment of immune therapies of HCC, further investigations are still required in this field.


Subject(s)
Carcinoma, Hepatocellular/therapy , Fibrosis/therapy , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy , Liver Neoplasms/therapy , Stem Cell Niche/drug effects , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Fibrosis/immunology , Fibrosis/pathology , Humans , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Stem Cell Niche/immunology
2.
Front Immunol ; 12: 784691, 2021.
Article in English | MEDLINE | ID: mdl-34956214

ABSTRACT

B-cell non-Hodgkin lymphoma (B-NHL) evolution and treatment are complicated by a high prevalence of relapses primarily due to the ability of malignant B cells to interact with tumor-supportive lymph node (LN) and bone marrow (BM) microenvironments. In particular, progressive alterations of BM stromal cells sustain the survival, proliferation, and drug resistance of tumor B cells during diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), and chronic lymphocytic leukemia (CLL). The current review describes how the crosstalk between BM stromal cells and lymphoma tumor cells triggers the establishment of the tumor supportive niche. DLBCL, FL, and CLL display distinct patterns of BM involvement, but in each case tumor-infiltrating stromal cells, corresponding to cancer-associated fibroblasts, exhibit specific phenotypic and functional features promoting the recruitment, adhesion, and survival of tumor cells. Tumor cell-derived extracellular vesicles have been recently proposed as playing a central role in triggering initial induction of tumor-supportive niches, notably within the BM. Finally, the disruption of the BM stroma reprogramming emerges as a promising therapeutic option in B-cell lymphomas. Targeting the crosstalk between BM stromal cells and malignant B cells, either through the inhibition of stroma-derived B-cell growth factors or through the mobilization of clonal B cells outside their supportive BM niche, should in particular be further evaluated as a way to avoid relapses by abrogating resistance niches.


Subject(s)
B-Lymphocytes/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphoma, Follicular/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , Mesenchymal Stem Cells/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Bone Marrow/pathology , Cell Communication/immunology , Cell Differentiation/immunology , Coculture Techniques , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma, Follicular/pathology , Lymphoma, Large B-Cell, Diffuse/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Stem Cell Niche/immunology , Tumor Cells, Cultured , Tumor Microenvironment/immunology
4.
Int Immunol ; 33(12): 821-826, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34668936

ABSTRACT

Most lineages of blood cells, including immune cells, are generated from hematopoietic stem cells (HSCs) in bone marrow throughout adult life. Since HSCs cannot expand on their own, they require and contact the special microenvironments, termed niches for their maintenance. HSC niches comprise supportive cells that provide adjacent HSCs with critical signals, including cytokines. Although bone marrow microenvironments have been thought to be complex, recent studies have demonstrated that the bone marrow-specific population of fibroblastic reticular cells with long processes, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells, which overlap strongly with leptin receptor (LepR)-expressing (LepR+) cells, is the major cellular component of niches for HSCs and lymphoid progenitors. CAR cells have salient features, expressing much higher levels of critical HSC niche factors than any other cell populations and function as self-renewing mesenchymal stem cells. Human counterpart of CAR cells is present and affected in diseases, including leukemia. Foxl1+ telocytes recently identified as the niche for intestinal stem cells share some features with CAR cells, suggesting that CAR cells might serve as a prototype for fibroblastic reticular cells creating niche for long-lived cells, including tissue stem cells and memory lymphocytes. These findings provided the basis for future mechanistic studies on the cross-talk between hematopoietic cells and microenvironments in both health and disease.


Subject(s)
Bone Marrow/immunology , Fibroblasts/immunology , Hematopoietic Stem Cells/immunology , Stem Cell Niche/immunology , Animals , Humans
5.
Front Immunol ; 12: 719349, 2021.
Article in English | MEDLINE | ID: mdl-34484226

ABSTRACT

In adult mammals, blood cells are formed from hematopoietic stem progenitor cells, which are controlled by a complex cellular microenvironment called "niche". Drosophila melanogaster is a powerful model organism to decipher the mechanisms controlling hematopoiesis, due both to its limited number of blood cell lineages and to the conservation of genes and signaling pathways throughout bilaterian evolution. Insect blood cells or hemocytes are similar to the mammalian myeloid lineage that ensures innate immunity functions. Like in vertebrates, two waves of hematopoiesis occur in Drosophila. The first wave takes place during embryogenesis. The second wave occurs at larval stages, where two distinct hematopoietic sites are identified: subcuticular hematopoietic pockets and a specialized hematopoietic organ called the lymph gland. In both sites, hematopoiesis is regulated by distinct niches. In hematopoietic pockets, sensory neurons of the peripheral nervous system provide a microenvironment that promotes embryonic hemocyte expansion and differentiation. In the lymph gland blood cells are produced from hematopoietic progenitors. A small cluster of cells called Posterior Signaling Centre (PSC) and the vascular system, along which the lymph gland develops, act collectively as a niche, under homeostatic conditions, to control the balance between maintenance and differentiation of lymph gland progenitors. In response to an immune stress such as wasp parasitism, lymph gland hematopoiesis is drastically modified and shifts towards emergency hematopoiesis, leading to increased progenitor proliferation and their differentiation into lamellocyte, a specific blood cell type which will neutralize the parasite. The PSC is essential to control this emergency response. In this review, we summarize Drosophila cellular and molecular mechanisms involved in the communication between the niche and hematopoietic progenitors, both under homeostatic and stress conditions. Finally, we discuss similarities between mechanisms by which niches regulate hematopoietic stem/progenitor cells in Drosophila and mammals.


Subject(s)
Cell Communication , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeostasis , Stem Cell Niche , Stress, Physiological , Animals , Cellular Microenvironment , Drosophila , Hemocytes/cytology , Hemocytes/metabolism , Larva , Models, Biological , Neurons/cytology , Neurons/metabolism , Stem Cell Niche/immunology , Stress, Physiological/immunology
6.
J Histochem Cytochem ; 69(12): 835-847, 2021 12.
Article in English | MEDLINE | ID: mdl-34165363

ABSTRACT

Cancer stem cells (CSCs) drive metastasis, treatment resistance, and tumor recurrence. CSCs reside within a niche, an anatomically distinct site within the tumor microenvironment (TME) that consists of malignant and non-malignant cells, including immune cells. The renin-angiotensin system (RAS), a critical regulator of stem cells and key developmental processes, plays a vital role in the TME. Non-malignant cells within the CSC niche and stem cell signaling pathways such as the Wnt, Hedgehog, and Notch pathways influence CSCs. Components of the RAS and cathepsins B and D that constitute bypass loops of the RAS are expressed on CSCs in many cancer types. There is extensive in vitro and in vivo evidence showing that RAS inhibition reduces tumor growth, cell proliferation, invasion, and metastasis. However, there is inconsistent epidemiological data on the effect of RAS inhibitors on cancer incidence and survival outcomes, attributed to different patient characteristics and methodologies used between studies. Further mechanistic studies are warranted to investigate the precise effects of the RAS on CSCs directly and/or the CSC niche. Targeting the RAS, its bypass loops, and convergent signaling pathways participating in the TME and other key stem cell pathways that regulate CSCs may be a novel approach to cancer treatment.


Subject(s)
Neoplasms/therapy , Neoplastic Stem Cells/metabolism , Renin-Angiotensin System/immunology , Stem Cell Niche/immunology , Animals , Cathepsins/immunology , Cell Proliferation , Drug Repositioning , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Humans , Signal Transduction , Tumor Microenvironment
7.
J Immunol ; 206(12): 3053-3063, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34078710

ABSTRACT

Systemic transplantation of stem cells from human exfoliated deciduous teeth (SHED) is used to treat systemic lupus erythematosus (SLE)-like disorders in MRL/lpr mice. However, the mechanisms underlying the SHED-based therapy remain unclear. In this study, we hypothesized that trophic factors within SHED-releasing extracellular vesicles (SHED-EVs) ameliorate the SLE-like phenotypes in MRL/lpr mice. SHED-EVs were isolated from the culture supernatant of SHED. SHED-EVs were treated with or without RNase and systemically administered to MRL/lpr mice. Subsequently, recipient bone marrow mesenchymal stem cells (BMMSCs) isolated from SHED-EV-administered MRL/lpr mice were examined for the in vitro and in vivo activity of hematopoietic niche formation and immunoregulation. Furthermore, the recipient BMMSCs were secondarily transplanted into MRL/lpr mice. The systemic SHED-EV infusion ameliorated the SLE-like phenotypes in MRL/lpr mice and improved the functions of recipient BMMSCs by rescuing Tert mRNA-associated telomerase activity, hematopoietic niche formation, and immunoregulation. The secondary transplantation of recipient BMMSCs recovered the immune condition and renal functions of MRL/lpr mice. The RNase treatment depleted RNAs, such as microRNAs, within SHED-EVs, and the RNA-depleted SHED-EVs attenuated the benefits of SHED-EVs in MRL/lpr mice. Collectively, our findings suggest that SHED-secreted RNAs, such as microRNAs, play a crucial role in treating SLE by targeting the telomerase activity of recipient BMMSCs.


Subject(s)
Extracellular Vesicles/immunology , Lupus Erythematosus, Systemic/immunology , Stem Cell Niche/immunology , Stem Cells/immunology , Telomerase/immunology , Tooth, Deciduous/immunology , Animals , Cells, Cultured , Child , Child, Preschool , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Inbred NOD , Mice, SCID
8.
Int J Mol Sci ; 22(8)2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33920983

ABSTRACT

Ovarian cancer is an aggressive gynaecological cancer with extremely poor prognosis, due to late diagnosis as well as the development of chemoresistance after first-line therapy. Research advances have found stem-like cells present in ovarian tumours, which exist in a dynamic niche and persist through therapy. The stem cell niche interacts extensively with the immune and non-immune components of the tumour microenvironment. Significant pathways associated with the cancer stem cell niche have been identified which interfere with the immune component of the tumour microenvironment, leading to immune surveillance evasion, dysfunction and suppression. This review aims to summarise current evidence-based knowledge on the cancer stem cell niche within the ovarian cancer tumour microenvironment and its effect on immune surveillance. Furthermore, the review seeks to understand the clinical consequences of this dynamic interaction by highlighting current therapies which target these processes.


Subject(s)
Immunologic Surveillance , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Stem Cell Niche/immunology , Animals , Female , Humans , Inflammation/pathology , Ovarian Neoplasms/therapy , Signal Transduction
9.
Front Immunol ; 12: 631279, 2021.
Article in English | MEDLINE | ID: mdl-33790904

ABSTRACT

Tissue engineering opens multiple opportunities in regenerative medicine, drug testing, and modeling of the hematopoiesis in health and disease. Recapitulating the organization of physiological microenvironments supporting leukocyte development is essential to model faithfully the development of immune cells. Hematopoietic organs are shaped by spatially organized niches defined by multiple cellular contributions. A shared feature of immune niches is the presence of mesenchymal stromal cells endowed with unique roles in organizing niche development, maintenance, and function. Here, we review challenges and opportunities in harnessing stromal cells for the engineering of artificial immune niches and hematopoietic organoids recapitulating leukocyte ontogeny both in vitro and in vivo.


Subject(s)
Mesenchymal Stem Cells/physiology , Stem Cell Niche/physiology , Stromal Cells/metabolism , Tissue Engineering/methods , Animals , Bone Marrow Cells/metabolism , Humans , Mesenchymal Stem Cells/immunology , Mice , Stem Cell Niche/genetics , Stem Cell Niche/immunology , Stromal Cells/immunology
10.
J Clin Invest ; 131(6)2021 03 15.
Article in English | MEDLINE | ID: mdl-33720051

ABSTRACT

Many solid cancers metastasize to the bone and bone marrow (BM). This process may occur even before the diagnosis of primary tumors, as evidenced by the discovery of disseminated tumor cells (DTCs) in patients without occult malignancies. The cellular fates and metastatic progression of DTCs are determined by complicated interactions between cancer cells and BM niches. Not surprisingly, these niches also play important roles in normal biology, including homeostasis and turnover of skeletal and hematopoiesis systems. In this Review, we summarize recent findings on functions of BM niches in bone metastasis (BoMet), particularly during the early stage of colonization. In light of the rich knowledge of hematopoiesis and osteogenesis, we highlight how DTCs may progress into overt BoMet by taking advantage of niche cells and their activities in tissue turnover, especially those related to immunomodulation and bone repair.


Subject(s)
Bone Neoplasms/secondary , Bone Marrow Neoplasms/immunology , Bone Marrow Neoplasms/pathology , Bone Marrow Neoplasms/secondary , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Remodeling/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Disease Progression , Female , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Humans , Immune Privilege , Immune Tolerance , Male , Models, Biological , Myeloid Cells/immunology , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neoplasm Metastasis/therapy , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Stem Cell Niche/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology
11.
FASEB J ; 35(1): e21234, 2021 01.
Article in English | MEDLINE | ID: mdl-33337557

ABSTRACT

Emerging evidences highlight importance of epigenetic regulation and their integration with transcriptional and cell signaling machinery in determining tissue resident adult pluripotent mesenchymal stem/stromal cell (MSC) activity, lineage commitment, and multicellular development. Histone modifying enzymes and large multi-subunit chromatin remodeling complexes and their cell type-specific plasticity remain the central defining features of gene regulation and establishment of tissue identity. Modulation of transcription factor expression gradient ex vivo and concomitant flexibility of higher order chromatin architecture in response to signaling cues are exciting approaches to regulate MSC activity and tissue rejuvenation. Being an important constituent of the adult bone marrow microenvironment/niche, pathophysiological perturbation in MSC homeostasis also causes impaired hematopoietic stem/progenitor cell function in a non-cell autonomous mechanism. In addition, pluripotent MSCs can function as immune regulatory cells, and they reside at the crossroad of innate and adaptive immune response pathways. Research in the past few years suggest that MSCs/stromal fibroblasts significantly contribute to the establishment of immunosuppressive microenvironment in shaping antitumor immunity. Therefore, it is important to understand mesenchymal stromal epigenome and transcriptional regulation to leverage its applications in regenerative medicine, epigenetic memory-guided trained immunity, immune-metabolic rewiring, and precision immune reprogramming. In this review, we highlight the latest developments and prospects in chromatin biology in determining MSC function in the context of lineage commitment and immunomodulation.


Subject(s)
Chromatin Assembly and Disassembly/immunology , Hematopoietic Stem Cells/immunology , Histones/immunology , Mesenchymal Stem Cells/immunology , Protein Processing, Post-Translational/immunology , Stem Cell Niche/immunology , Animals , Hematopoietic Stem Cells/cytology , Humans , Mesenchymal Stem Cells/cytology
12.
Front Immunol ; 11: 600127, 2020.
Article in English | MEDLINE | ID: mdl-33324418

ABSTRACT

Studies over the last couple of decades have shown that hematopoietic stem cells (HSCs) are critically dependent on cytokines such as Stem Cell Factor and other signals provided by bone marrow niches comprising of mesenchymal stem and progenitor cells (MSPCs) and endothelial cells (ECs). Because of their critical roles in HSC maintenance the niches formed by MSPCs and ECs are commonly referred to as HSC niches. For the most part, the signals required for HSC maintenance act in a short-range manner, which imposes the necessity for directional and positional cues in order for HSCs to localize and be retained properly in stem cell niches. The chemokine CXCL12 and its Gαi protein coupled receptor CXCR4, besides promoting HSC quiescence directly, also play instrumental roles in enabling HSCs to access bone marrow stem cell niches. Recent studies have revealed, however, that HSC niches also provide a constellation of hematopoietic cytokines that are critical for the production of most, if not all, blood cell types. Some hematopoietic cytokines, namely IL-7 and IL-15 produced by HSC niches, are not only required for lymphopoiesis but are also essential for memory T cell maintenance. Consequently, hematopoietic progenitors and differentiated immune cells, such as memory T cell subsets, also depend on the CXCL12/CXCR4 axis for migration into bone marrow and interactions with MSPCs and ECs. Similarly, subsets of antibody-secreting plasma cells also reside in close association with CXCL12-producing MSPCs in the bone marrow and require the CXCR4/CXCL12 axis for survival and long-term maintenance. Collectively, these studies demonstrate a broad range of key physiological roles, spanning blood cell production and maintenance of immunological memory, that are orchestrated by stem cell niches through a common and simple mechanism: CXCL12/CXCR4-mediated cell recruitment followed by receipt of a maintenance and/or instructive signal. A fundamental flaw of this type of cellular organization is revealed by myeloid and lymphoid leukemias, which target stem cell niches and induce profound transcriptomic changes that result in reduced hematopoietic activity and altered mesenchymal cell differentiation.


Subject(s)
Hematopoietic Stem Cells/immunology , Immunologic Memory , Signal Transduction/immunology , Stem Cell Niche/immunology , Animals , Endothelial Cells/immunology , Humans , Mesenchymal Stem Cells/immunology
13.
Nat Commun ; 11(1): 3062, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32546788

ABSTRACT

Anti-tuberculosis (TB) drugs, while being highly potent in vitro, require prolonged treatment to control Mycobacterium tuberculosis (Mtb) infections in vivo. We report here that mesenchymal stem cells (MSCs) shelter Mtb to help tolerate anti-TB drugs. MSCs readily take up Mtb and allow unabated mycobacterial growth despite having a functional innate pathway of phagosome maturation. Unlike macrophage-resident ones, MSC-resident Mtb tolerates anti-TB drugs remarkably well, a phenomenon requiring proteins ABCC1, ABCG2 and vacuolar-type H+ATPases. Additionally, the classic pro-inflammatory cytokines IFNγ and TNFα aid mycobacterial growth within MSCs. Mechanistically, evading drugs and inflammatory cytokines by MSC-resident Mtb is dependent on elevated PGE2 signaling, which we verify in vivo analyzing sorted CD45-Sca1+CD73+-MSCs from lungs of infected mice. Moreover, MSCs are observed in and around human tuberculosis granulomas, harboring Mtb bacilli. We therefore propose, targeting the unique immune-privileged niche, provided by MSCs to Mtb, can have a major impact on tuberculosis prevention and cure.


Subject(s)
Antitubercular Agents/pharmacology , Mesenchymal Stem Cells/microbiology , Mycobacterium tuberculosis/pathogenicity , Stem Cell Niche/immunology , Tuberculosis/microbiology , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Cells, Cultured , Dinoprostone/metabolism , Host-Pathogen Interactions , Humans , Interferon-gamma/pharmacology , Isoniazid/pharmacology , Lysosomes/microbiology , Mesenchymal Stem Cells/drug effects , Mice, Inbred C57BL , Multidrug Resistance-Associated Proteins/metabolism , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , Neoplasm Proteins/metabolism , Phagosomes/microbiology , Tuberculosis/pathology , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/pathology , Tumor Necrosis Factor-alpha/pharmacology
14.
Mater Sci Eng C Mater Biol Appl ; 109: 110508, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32228925

ABSTRACT

The control of early inflammatory reactions and recruitment of progenitor cells are critical for subsequent tissue repair and regeneration after biomaterial implantation. The aim of this study was to design a multi-functional biomaterial with a controlled drug delivery system to create an optimal local environment for early osteogenesis. Here, the anti-inflammatory cytokine IL-4 and pro-osteogenic RGD peptide were assembled layer-by-layer on TiO2 nanotubes. A poly(dopamine) (DOP) coating was employed onto TiO2 nanotubes (T/DOP) to functionalized with IL-4 (T/DOP-IL4). Then, a carboxymethyl chitosan hydrogel layer (CG) was generated on T/DOP-IL4 to control IL-4 release and RGD peptide immobilization. Cell co-culture models were applied to study macrophage polarization on various material surfaces and the regulation of mesenchymal stromal cell (MSC) osteogenic differentiation. Our data suggest that T/DOP-IL4/CG-RGD surfaces developed in this study are multi-functional, and can not only drive phenotypic changes in macrophages (switching to anti-inflammatory M2 phenotype), resulting in the production of reparative cytokines such as IL-10, but also enhance MSC differentiation related to the activation of BMP/SMAD/RUNX2 signaling. This study further confirmed that the introduction of anti-inflammatory cytokine (IL-4) and cell adhesive motif (RGD) onto Ti substrate can work synergistically to generate a more favorable early-stage osteo-immune environment with superior osteogenic properties, thus representing a potential ideal surface for the generation of bone biomaterials.


Subject(s)
Hydrogels , Interleukin-4 , Mesenchymal Stem Cells/immunology , Nanotubes/chemistry , Oligopeptides , Stem Cell Niche/immunology , Titanium , Animals , Coculture Techniques , Hydrogels/chemistry , Hydrogels/pharmacology , Interleukin-4/chemistry , Interleukin-4/pharmacology , Macrophages/immunology , Mice , Oligopeptides/chemistry , Oligopeptides/pharmacology , Osteogenesis/drug effects , Osteogenesis/immunology , RAW 264.7 Cells , Stem Cell Niche/drug effects , Titanium/chemistry , Titanium/pharmacology
15.
Med Sci (Paris) ; 36(1): 69-72, 2020 Jan.
Article in French | MEDLINE | ID: mdl-32014101

ABSTRACT

TITLE: Le dialogue entre les cellules souches intestinales et les lymphocytes T CD4+ module l'homéostasie des cellules souches - Module d'immunologie virologie et cancer du Master de cancérologie de Lyon. ABSTRACT: Dans le cadre d'un partenariat avec médecine/sciences, et pour la troisième année, des étudiants du module d'immunologie virologie et cancer du Master de cancérologie de Lyon présentent une analyse d'articles scientifiques récents faisant état d'observations innovantes et importantes. Ce travail a été encadré par des chercheurs confirmés du département d'immunologie, virologie et inflammation du CRCL. Le master de cancérologie de Lyon (Lyon1-VetAgroSup) accueille chaque année 30 à 40 étudiants en M1 et en M2. Ce master dit « d'excellence ¼ assure aux étudiants de M1 une formation à la cancérologie reposant sur un socle de base commun (biologie cellulaire, moléculaire, immunologie, bio-statistique...). En M2, les étudiants peuvent choisir l'une des trois spécialités suivantes : le Master recherche « Recherche en cancérologie ¼, le Master recherche et professionnel « Technologie haut débit en cancérologie ¼ et enfin le Master recherche et professionnel « Innovations thérapeutiques en cancérologie ¼. Le Master de cancérologie de Lyon repose sur une forte implication des chercheurs et enseignants-chercheurs du laboratoire d'excellence en développement et cancérologie (LabEx DEVweCAN), ainsi que sur un partenariat solide avec plusieurs instituts dont le MIT (Massachusetts Institute of Technology, Cambridge, États-Unis), l'université d'Harvard (Boston, États-Unis), l'université Johns Hopkins (Baltimore, États-Unis), l'Imperial College of London (Royaume-Uni), les universités de Jiao Tong (République Populaire de Chine) et de Tokyo (Japon), entre autres. Pour plus d'information : http://devwecan.universite-lyon.fr/formation/.


Subject(s)
Adult Stem Cells/physiology , CD4-Positive T-Lymphocytes/physiology , Cell Communication/physiology , Intestinal Mucosa/cytology , Animals , Homeostasis/physiology , Humans , Intestinal Mucosa/immunology , Intestines/cytology , Intestines/immunology , Stem Cell Niche/immunology
16.
Nature ; 576(7787): 465-470, 2019 12.
Article in English | MEDLINE | ID: mdl-31827286

ABSTRACT

Tumour-infiltrating lymphocytes are associated with a survival benefit in several tumour types and with the response to immunotherapy1-8. However, the reason some tumours have high CD8 T cell infiltration while others do not remains unclear. Here we investigate the requirements for maintaining a CD8 T cell response against human cancer. We find that CD8 T cells within tumours consist of distinct populations of terminally differentiated and stem-like cells. On proliferation, stem-like CD8 T cells give rise to more terminally differentiated, effector-molecule-expressing daughter cells. For many T cells to infiltrate the tumour, it is critical that this effector differentiation process occur. In addition, we show that these stem-like T cells reside in dense antigen-presenting-cell niches within the tumour, and that tumours that fail to form these structures are not extensively infiltrated by T cells. Patients with progressive disease lack these immune niches, suggesting that niche breakdown may be a key mechanism of immune escape.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , Stem Cells/cytology , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/metabolism , Disease Progression , Epigenesis, Genetic , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Neoplasms/pathology , Stem Cell Niche/immunology , Transcription, Genetic , Tumor Escape/genetics , Tumor Escape/immunology
17.
Trans Am Clin Climatol Assoc ; 130: 246-254, 2019.
Article in English | MEDLINE | ID: mdl-31516189

ABSTRACT

A major limitation of current leukemia treatment is that most patients ultimately relapse. Leukemia cells show heterogeneous potential and response to treatment. We have shown that primitive leukemia stem cells (LSC) in chronic myelogenous leukemia resist elimination by treatment, and persist as a source of relapse. The bone marrow microenvironment (BMM) plays a critical role in of hematopoietic stem cell maintenance and regulation. There is increasing interest in the role of the BMM in promoting LSC maintenance, resistance to therapy, and ultimately disease relapse. Recent studies have shown that leukemia-induced changes in the BMM provide a competitive growth advantage to LSC, and support their preservation after treatment. We are studying mechanisms of niche regulation of LSC to guide development of novel approaches to target LSC and enhance cures.


Subject(s)
Drug Resistance, Neoplasm/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Neoplastic Stem Cells/physiology , Protein Kinase Inhibitors/therapeutic use , Stem Cell Niche/physiology , Tumor Microenvironment/physiology , Apoptosis , Bone Marrow , Cell Proliferation , Cytokines/immunology , Drug Resistance, Neoplasm/immunology , Fusion Proteins, bcr-abl/antagonists & inhibitors , Hematopoietic Stem Cells , Humans , Neoplasm Recurrence, Local , Remission Induction , Stem Cell Niche/immunology , Tumor Microenvironment/immunology
18.
J Theor Biol ; 482: 109999, 2019 12 07.
Article in English | MEDLINE | ID: mdl-31493486

ABSTRACT

Accumulating experimental and clinical evidence suggest that the immune response to cancer is not exclusively anti-tumor. Indeed, the pro-tumor roles of the immune system  -  as suppliers of growth and pro-angiogenic factors or defenses against cytotoxic immune attacks, for example  -  have been long appreciated, but relatively few theoretical works have considered their effects. Inspired by the recently proposed "immune-mediated" theory of metastasis, we develop a mathematical model for tumor-immune interactions at two anatomically distant sites, which includes both anti- and pro-tumor immune effects, and the experimentally observed tumor-induced phenotypic plasticity of immune cells (tumor "education" of the immune cells). Upon confrontation of our model to experimental data, we use it to evaluate the implications of the immune-mediated theory of metastasis. We find that tumor education of immune cells may explain the relatively poor performance of immunotherapies, and that many metastatic phenomena, including metastatic blow-up, dormancy, and metastasis to sites of injury, can be explained by the immune-mediated theory of metastasis. Our results suggest that further work is warranted to fully elucidate the pro-tumor effects of the immune system in metastatic cancer.


Subject(s)
Immune System/physiology , Immunotherapy/adverse effects , Models, Theoretical , Neoplasm Metastasis/immunology , Tumor Escape/immunology , Cell Proliferation/physiology , Humans , Immunotherapy/methods , Neoplasm Invasiveness/immunology , Neoplasm Metastasis/pathology , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Neoplastic Stem Cells/physiology , Stem Cell Niche/immunology , Tumor Burden/immunology , Tumor Microenvironment/immunology
19.
Nature ; 572(7771): 603-608, 2019 08.
Article in English | MEDLINE | ID: mdl-31462798

ABSTRACT

Direct investigation of the early cellular changes induced by metastatic cells within the surrounding tissue remains a challenge. Here we present a system in which metastatic cancer cells release a cell-penetrating fluorescent protein, which is taken up by neighbouring cells and enables spatial identification of the local metastatic cellular environment. Using this system, tissue cells with low representation in the metastatic niche can be identified and characterized within the bulk tissue. To highlight its potential, we applied this strategy to study the cellular environment of metastatic breast cancer cells in the lung. We report the presence of cancer-associated parenchymal cells, which exhibit stem-cell-like features, expression of lung progenitor markers, multi-lineage differentiation potential and self-renewal activity. In ex vivo assays, lung epithelial cells acquire a cancer-associated parenchymal-cell-like phenotype when co-cultured with cancer cells and support their growth. These results highlight the potential of this method as a platform for new discoveries.


Subject(s)
Cell Lineage , Cell Tracking/methods , Neoplasm Metastasis/pathology , Neoplastic Stem Cells/pathology , Parenchymal Tissue/pathology , Staining and Labeling/methods , Stem Cell Niche , Tumor Microenvironment , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Differentiation , Coculture Techniques , Epithelial Cells/pathology , Female , Humans , Luminescent Proteins/analysis , Luminescent Proteins/chemistry , Luminescent Proteins/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Male , Mice , Neoplasm Metastasis/immunology , Neutrophils/pathology , Organoids/pathology , Stem Cell Niche/immunology , Tumor Microenvironment/immunology , Red Fluorescent Protein
20.
Immunity ; 51(1): 90-103.e3, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31278057

ABSTRACT

The key sites within the gastrointestinal (GI) tract where T cells mediate effector responses and the impact of these responses on intestinal stem cells (ISCs) remain unclear. Using experimental bone marrow transplantation to model immune-mediated GI damage and 3D imaging to analyze T cell localization, we found that the ISC compartment is the primary intestinal site targeted by T cells after transplantation. Recruitment to the crypt base region resulted in direct T cell engagement with the stem cell compartment and loss of crypt base columnar ISCs, which expressed both MHC classes I and II. Vasculature expressing the adhesion molecule MAdCAM-1 clustered near the crypt base, preferentially regulating crypt compartment invasion and ISC reduction without affecting T cell migration to villi. These findings indicate that allogeneic T cells rapidly access the stem cell niche after transplantation, and this targeted recruitment to the stem cell compartment results in ISC loss during immune-mediated GI damage.


Subject(s)
Adult Stem Cells/immunology , Bone Marrow Transplantation , Intestinal Mucosa/immunology , Stem Cell Niche/immunology , T-Lymphocytes/immunology , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Cell Adhesion Molecules/metabolism , Cell Movement , Cytotoxicity, Immunologic , Female , Humans , Imaging, Three-Dimensional , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Models, Animal , Mucoproteins , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...