Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Int Immunol ; 36(7): 339-352, 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38430523

RESUMEN

Bone marrow is a dynamic organ composed of stem cells that constantly receive signals from stromal cells and other hematopoietic cells in the niches of the bone marrow to maintain hematopoiesis and generate immune cells. Perturbation of the bone marrow microenvironment by infection and inflammation affects hematopoiesis and may affect immune cell development. Little is known about the effect of malaria on the bone marrow stromal cells that govern the hematopoietic stem cell (HSC) niche. In this study, we demonstrate that the mesenchymal stromal CXCL12-abundant reticular (CAR) cell population is reduced during acute malaria infection. The reduction of CXCL12 and interleukin-7 signals in the bone marrow impairs the lymphopoietic niche, leading to the depletion of common lymphoid progenitors, B cell progenitors, and mature B cells, including plasma cells in the bone marrow. We found that interferon-γ (IFNγ) is responsible for the upregulation of Sca1 on CAR cells, yet the decline in CAR cell and B cell populations in the bone marrow is IFNγ-independent. In contrast to the decline in B cell populations, HSCs and multipotent progenitors increased with the expansion of myelopoiesis and erythropoiesis, indicating a bias in the differentiation of multipotent progenitors during malaria infection. These findings suggest that malaria may affect host immunity by modulating the bone marrow niche.


Asunto(s)
Linfocitos B , Médula Ósea , Quimiocina CXCL12 , Malaria , Ratones Endogámicos C57BL , Animales , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/inmunología , Ratones , Malaria/inmunología , Malaria/parasitología , Linfocitos B/inmunología , Médula Ósea/inmunología , Médula Ósea/parasitología , Nicho de Células Madre/inmunología , Interferón gamma/metabolismo , Interferón gamma/inmunología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo
2.
J Histochem Cytochem ; 70(1): 53-81, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34751050

RESUMEN

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.


Asunto(s)
Carcinoma Hepatocelular/terapia , Fibrosis/terapia , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia , Neoplasias Hepáticas/terapia , Nicho de Células Madre/efectos de los fármacos , Animales , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Fibrosis/inmunología , Fibrosis/patología , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Nicho de Células Madre/inmunología
3.
Front Immunol ; 12: 784691, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34956214

RESUMEN

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.


Asunto(s)
Linfocitos B/inmunología , Leucemia Linfocítica Crónica de Células B/inmunología , Linfoma Folicular/inmunología , Linfoma de Células B Grandes Difuso/inmunología , Células Madre Mesenquimatosas/inmunología , Linfocitos B/metabolismo , Linfocitos B/patología , Médula Ósea/patología , Comunicación Celular/inmunología , Diferenciación Celular/inmunología , Técnicas de Cocultivo , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/metabolismo , Humanos , Leucemia Linfocítica Crónica de Células B/patología , Linfoma Folicular/patología , Linfoma de Células B Grandes Difuso/patología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Nicho de Células Madre/inmunología , Células Tumorales Cultivadas , Microambiente Tumoral/inmunología
5.
Int Immunol ; 33(12): 821-826, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34668936

RESUMEN

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.


Asunto(s)
Médula Ósea/inmunología , Fibroblastos/inmunología , Células Madre Hematopoyéticas/inmunología , Nicho de Células Madre/inmunología , Animales , Humanos
6.
Front Immunol ; 12: 719349, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34484226

RESUMEN

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.


Asunto(s)
Comunicación Celular , Hematopoyesis , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Nicho de Células Madre , Estrés Fisiológico , Animales , Microambiente Celular , Drosophila , Hemocitos/citología , Hemocitos/metabolismo , Larva , Modelos Biológicos , Neuronas/citología , Neuronas/metabolismo , Nicho de Células Madre/inmunología , Estrés Fisiológico/inmunología
7.
J Histochem Cytochem ; 69(12): 835-847, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34165363

RESUMEN

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.


Asunto(s)
Neoplasias/terapia , Células Madre Neoplásicas/metabolismo , Sistema Renina-Angiotensina/inmunología , Nicho de Células Madre/inmunología , Animales , Catepsinas/inmunología , Proliferación Celular , Reposicionamiento de Medicamentos , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Humanos , Transducción de Señal , Microambiente Tumoral
8.
J Immunol ; 206(12): 3053-3063, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34078710

RESUMEN

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.


Asunto(s)
Vesículas Extracelulares/inmunología , Lupus Eritematoso Sistémico/inmunología , Nicho de Células Madre/inmunología , Células Madre/inmunología , Telomerasa/inmunología , Diente Primario/inmunología , Animales , Células Cultivadas , Niño , Preescolar , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Ratones Endogámicos NOD , Ratones SCID
9.
Front Immunol ; 12: 631279, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33790904

RESUMEN

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.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Nicho de Células Madre/fisiología , Células del Estroma/metabolismo , Ingeniería de Tejidos/métodos , Animales , Células de la Médula Ósea/metabolismo , Humanos , Células Madre Mesenquimatosas/inmunología , Ratones , Nicho de Células Madre/genética , Nicho de Células Madre/inmunología , Células del Estroma/inmunología
10.
Int J Mol Sci ; 22(8)2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33920983

RESUMEN

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.


Asunto(s)
Vigilancia Inmunológica , Células Madre Neoplásicas/patología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Nicho de Células Madre/inmunología , Animales , Femenino , Humanos , Inflamación/patología , Neoplasias Ováricas/terapia , Transducción de Señal
11.
J Clin Invest ; 131(6)2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33720051

RESUMEN

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.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Médula Ósea/inmunología , Neoplasias de la Médula Ósea/patología , Neoplasias de la Médula Ósea/secundario , Neoplasias Óseas/inmunología , Neoplasias Óseas/patología , Remodelación Ósea/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Progresión de la Enfermedad , Femenino , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/patología , Humanos , Privilegio Inmunológico , Tolerancia Inmunológica , Masculino , Modelos Biológicos , Células Mieloides/inmunología , Metástasis de la Neoplasia/inmunología , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/terapia , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Nicho de Células Madre/inmunología , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/inmunología
12.
FASEB J ; 35(1): e21234, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33337557

RESUMEN

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.


Asunto(s)
Ensamble y Desensamble de Cromatina/inmunología , Células Madre Hematopoyéticas/inmunología , Histonas/inmunología , Células Madre Mesenquimatosas/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Nicho de Células Madre/inmunología , Animales , Células Madre Hematopoyéticas/citología , Humanos , Células Madre Mesenquimatosas/citología
13.
Front Immunol ; 11: 600127, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33324418

RESUMEN

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.


Asunto(s)
Células Madre Hematopoyéticas/inmunología , Memoria Inmunológica , Transducción de Señal/inmunología , Nicho de Células Madre/inmunología , Animales , Células Endoteliales/inmunología , Humanos , Células Madre Mesenquimatosas/inmunología
14.
Nat Commun ; 11(1): 3062, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32546788

RESUMEN

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.


Asunto(s)
Antituberculosos/farmacología , Células Madre Mesenquimatosas/microbiología , Mycobacterium tuberculosis/patogenicidad , Nicho de Células Madre/inmunología , Tuberculosis/microbiología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Animales , Células Cultivadas , Dinoprostona/metabolismo , Interacciones Huésped-Patógeno , Humanos , Interferón gamma/farmacología , Isoniazida/farmacología , Lisosomas/microbiología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos C57BL , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/crecimiento & desarrollo , Proteínas de Neoplasias/metabolismo , Fagosomas/microbiología , Tuberculosis/patología , Tuberculosis Pulmonar/tratamiento farmacológico , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología , Factor de Necrosis Tumoral alfa/farmacología
15.
Mater Sci Eng C Mater Biol Appl ; 109: 110508, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32228925

RESUMEN

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.


Asunto(s)
Hidrogeles , Interleucina-4 , Células Madre Mesenquimatosas/inmunología , Nanotubos/química , Oligopéptidos , Nicho de Células Madre/inmunología , Titanio , Animales , Técnicas de Cocultivo , Hidrogeles/química , Hidrogeles/farmacología , Interleucina-4/química , Interleucina-4/farmacología , Macrófagos/inmunología , Ratones , Oligopéptidos/química , Oligopéptidos/farmacología , Osteogénesis/efectos de los fármacos , Osteogénesis/inmunología , Células RAW 264.7 , Nicho de Células Madre/efectos de los fármacos , Titanio/química , Titanio/farmacología
16.
Med Sci (Paris) ; 36(1): 69-72, 2020 Jan.
Artículo en Francés | MEDLINE | ID: mdl-32014101

RESUMEN

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/.


Asunto(s)
Células Madre Adultas/fisiología , Linfocitos T CD4-Positivos/fisiología , Comunicación Celular/fisiología , Mucosa Intestinal/citología , Animales , Homeostasis/fisiología , Humanos , Mucosa Intestinal/inmunología , Intestinos/citología , Intestinos/inmunología , Nicho de Células Madre/inmunología
17.
Nature ; 576(7787): 465-470, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31827286

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias/inmunología , Células Madre/citología , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/metabolismo , Progresión de la Enfermedad , Epigénesis Genética , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Humanos , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Neoplasias/patología , Nicho de Células Madre/inmunología , Transcripción Genética , Escape del Tumor/genética , Escape del Tumor/inmunología
18.
Trans Am Clin Climatol Assoc ; 130: 246-254, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31516189

RESUMEN

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.


Asunto(s)
Resistencia a Antineoplásicos/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Células Madre Neoplásicas/fisiología , Inhibidores de Proteínas Quinasas/uso terapéutico , Nicho de Células Madre/fisiología , Microambiente Tumoral/fisiología , Apoptosis , Médula Ósea , Proliferación Celular , Citocinas/inmunología , Resistencia a Antineoplásicos/inmunología , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Células Madre Hematopoyéticas , Humanos , Recurrencia Local de Neoplasia , Inducción de Remisión , Nicho de Células Madre/inmunología , Microambiente Tumoral/inmunología
19.
J Theor Biol ; 482: 109999, 2019 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-31493486

RESUMEN

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.


Asunto(s)
Sistema Inmunológico/fisiología , Inmunoterapia/efectos adversos , Modelos Teóricos , Metástasis de la Neoplasia/inmunología , Escape del Tumor/inmunología , Proliferación Celular/fisiología , Humanos , Inmunoterapia/métodos , Invasividad Neoplásica/inmunología , Metástasis de la Neoplasia/patología , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Células Madre Neoplásicas/fisiología , Nicho de Células Madre/inmunología , Carga Tumoral/inmunología , Microambiente Tumoral/inmunología
20.
Nature ; 572(7771): 603-608, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31462798

RESUMEN

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.


Asunto(s)
Linaje de la Célula , Rastreo Celular/métodos , Metástasis de la Neoplasia/patología , Células Madre Neoplásicas/patología , Tejido Parenquimatoso/patología , Coloración y Etiquetado/métodos , Nicho de Células Madre , Microambiente Tumoral , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Diferenciación Celular , Técnicas de Cocultivo , Células Epiteliales/patología , Femenino , Humanos , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/química , Proteínas Luminiscentes/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Masculino , Ratones , Metástasis de la Neoplasia/inmunología , Neutrófilos/patología , Organoides/patología , Nicho de Células Madre/inmunología , Microambiente Tumoral/inmunología , Proteína Fluorescente Roja
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA