Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Nat Commun ; 15(1): 3297, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38740748

RESUMEN

Despite abundant evidence demonstrating that platelets foster metastasis, anti-platelet agents have low therapeutic potential due to the risk of hemorrhages. In addition, whether platelets can regulate metastasis at the late stages of the disease remains unknown. In this study, we subject syngeneic models of metastasis to various thrombocytopenic regimes to show that platelets provide a biphasic contribution to metastasis. While potent intravascular binding of platelets to tumor cells efficiently promotes metastasis, platelets further support the outgrowth of established metastases via immune suppression. Genetic depletion and pharmacological targeting of the glycoprotein VI (GPVI) platelet-specific receptor in humanized mouse models efficiently reduce the growth of established metastases, independently of active platelet binding to tumor cells in the bloodstream. Our study demonstrates therapeutic efficacy when targeting animals bearing growing metastases. It further identifies GPVI as a molecular target whose inhibition can impair metastasis without inducing collateral hemostatic perturbations.


Asunto(s)
Plaquetas , Metástasis de la Neoplasia , Glicoproteínas de Membrana Plaquetaria , Animales , Plaquetas/metabolismo , Plaquetas/efectos de los fármacos , Humanos , Ratones , Glicoproteínas de Membrana Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/genética , Línea Celular Tumoral , Femenino , Ratones Endogámicos C57BL
2.
AIDS ; 37(1): 43-49, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36001527

RESUMEN

OBJECTIVES: Broadly neutralizing antibodies have been proposed as key actors for HIV vaccine development. However, they display features of highly matured antibodies, hampering their induction by vaccination. As protective broadly neutralizing antibodies should be induced rapidly after vaccination and should neutralize the early-transmitted founder (T/F) viruses, we searched whether such antibodies may be induced following HIV infection. DESIGN: Sera were collected during acute infection (Day 0) and at viral set point (Month 6/12) and the neutralizing activity against T/F strains was investigated. Neutralizing activity in sera collected from chronic progressor was analyzed in parallel. METHODS: We compared neutralizing activity against T/F strains with neutralizing activity against non-T/F strains using the conventional TZM-bL neutralizing assay. RESULTS: We found neutralizing antibodies (nAbs) preferentially directed against T/F viruses in sera collected shortly after infection. This humoral response evolved by shifting to nAbs directed against non-T/F strains. CONCLUSION: Although features associated with nAbs directed against T/F viruses need further investigations, these early-induced nAbs may display lesser maturation characteristics; therefore, this might increase their interest for future vaccine designs.


Asunto(s)
Infecciones por VIH , Humanos , Infecciones por VIH/prevención & control , Anticuerpos ampliamente neutralizantes
3.
Sci Transl Med ; 14(628): eabj7521, 2022 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-34698500

RESUMEN

The drivers of critical coronavirus disease 2019 (COVID-19) remain unknown. Given major confounding factors such as age and comorbidities, true mediators of this condition have remained elusive. We used a multi-omics analysis combined with artificial intelligence in a young patient cohort where major comorbidities were excluded at the onset. The cohort included 47 "critical" (in the intensive care unit under mechanical ventilation) and 25 "non-critical" (in a non-critical care ward) patients with COVID-19 and 22 healthy individuals. The analyses included whole-genome sequencing, whole-blood RNA sequencing, plasma and blood mononuclear cell proteomics, cytokine profiling, and high-throughput immunophenotyping. An ensemble of machine learning, deep learning, quantum annealing, and structural causal modeling were used. Patients with critical COVID-19 were characterized by exacerbated inflammation, perturbed lymphoid and myeloid compartments, increased coagulation, and viral cell biology. Among differentially expressed genes, we observed up-regulation of the metalloprotease ADAM9. This gene signature was validated in a second independent cohort of 81 critical and 73 recovered patients with COVID-19 and was further confirmed at the transcriptional and protein level and by proteolytic activity. Ex vivo ADAM9 inhibition decreased severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uptake and replication in human lung epithelial cells. In conclusion, within a young, otherwise healthy, cohort of individuals with COVID-19, we provide the landscape of biological perturbations in vivo where a unique gene signature differentiated critical from non-critical patients. We further identified ADAM9 as a driver of disease severity and a candidate therapeutic target.


Asunto(s)
COVID-19 , Proteínas ADAM , Inteligencia Artificial , Humanos , Unidades de Cuidados Intensivos , Proteínas de la Membrana , Respiración Artificial , SARS-CoV-2
4.
AIDS ; 36(4): 487-499, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-34581307

RESUMEN

OBJECTIVE: Spontaneous control of HIV replication without treatment in HIV-1 controllers (HICs) was associated with the development of an efficient T-cell response. In addition, increasing data suggest that the humoral response participates in viral clearance. DESIGN: In-depth characterization of Ab response in HICs may help to define new parameters associated with this control. METHODS: We assessed the levels of total and HIV-specific IgA and IgG subtypes induction and their functional potencies - that is, neutralization, phagocytosis, antibody-dependent cellular cytotoxicity (ADCC), according to the individual's major histocompatibility complex class I (HLA)-B∗57 status, and compared it with nontreated chronic progressors. RESULTS: We found that despite an undetectable viral load, HICs displayed HIV-specific IgG levels similar to those of chronic progressors. Interestingly, our compelling multifunctional analysis demonstrates that the functional Ab profile, by itself, allowed to discriminate HLA-B∗57+ HICs from HLA-B∗57- HICs and chronic progressors. CONCLUSION: These results show that HICs display a particular HIV-specific antibody (Ab) profile that may participate in HIV control and emphasize the relevance of multifunctional Ab response analysis in future Ab-driven vaccine studies.


Asunto(s)
Infecciones por VIH , VIH-1 , Anticuerpos Anti-VIH , VIH no-Progresivos , Antígenos HLA-B , Humanos , Inmunoglobulina G , Carga Viral
6.
Front Immunol ; 12: 636108, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34290694

RESUMEN

Radiotherapy, the most frequent treatment of oral squamous cell carcinomas (OSCC) besides surgery is employed to kill tumor cells but, radiotherapy may also promote tumor relapse where the immune-suppressive tumor microenvironment (TME) could be instrumental. We established a novel syngeneic grafting model from a carcinogen-induced tongue tumor, OSCC13, to address the impact of radiotherapy on OSCC. This model revealed similarities with human OSCC, recapitulating carcinogen-induced mutations found in smoking associated human tongue tumors, abundant tumor infiltrating leukocytes (TIL) and, spontaneous tumor cell dissemination to the local lymph nodes. Cultured OSCC13 cells and OSCC13-derived tongue tumors were sensitive to irradiation. At the chosen dose of 2 Gy mimicking treatment of human OSCC patients not all tumor cells were killed allowing to investigate effects on the TME. By investigating expression of the extracellular matrix molecule tenascin-C (TNC), an indicator of an immune suppressive TME, we observed high local TNC expression and TIL infiltration in the irradiated tumors. In a TNC knockout host the TME appeared less immune suppressive with a tendency towards more tumor regression than in WT conditions. Altogether, our novel syngeneic tongue OSCC grafting model, sharing important features with the human OSCC disease could be relevant for future anti-cancer targeting of OSCC by radiotherapy and other therapeutic approaches.


Asunto(s)
Ganglios Linfáticos/efectos de la radiación , Carcinoma de Células Escamosas de Cabeza y Cuello/radioterapia , Tenascina/metabolismo , Neoplasias de la Lengua/radioterapia , Animales , Línea Celular Tumoral , Femenino , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Metástasis Linfática , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Trasplante de Neoplasias , Tolerancia a Radiación , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/secundario , Tenascina/genética , Neoplasias de la Lengua/genética , Neoplasias de la Lengua/metabolismo , Neoplasias de la Lengua/patología , Trasplante Isogénico , Carga Tumoral/efectos de la radiación , Microambiente Tumoral
7.
Sci Rep ; 11(1): 13144, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162963

RESUMEN

Tumor progression and metastatic dissemination are driven by cell-intrinsic and biomechanical cues that favor the growth of life-threatening secondary tumors. We recently identified pro-metastatic vascular regions with blood flow profiles that are permissive for the arrest of circulating tumor cells. We have further established that such flow profiles also control endothelial remodeling, which favors extravasation of arrested CTCs. Yet, how shear forces control endothelial remodeling is unknown. In the present work, we aimed at dissecting the cellular and molecular mechanisms driving blood flow-dependent endothelial remodeling. Transcriptomic analysis of endothelial cells revealed that blood flow enhanced VEGFR signaling, among others. Using a combination of in vitro microfluidics and intravital imaging in zebrafish embryos, we now demonstrate that the early flow-driven endothelial response can be prevented upon specific inhibition of VEGFR tyrosine kinase and subsequent signaling. Inhibitory targeting of VEGFRs reduced endothelial remodeling and subsequent metastatic extravasation. These results confirm the importance of VEGFR-dependent endothelial remodeling as a driving force of CTC extravasation and metastatic dissemination. Furthermore, the present work suggests that therapies targeting endothelial remodeling might be a relevant clinical strategy in order to impede metastatic progression.


Asunto(s)
Endotelio Vascular/fisiología , Hemorreología , Migración Transendotelial y Transepitelial , Animales , Animales Modificados Genéticamente , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Embrión no Mamífero/irrigación sanguínea , Embrión no Mamífero/fisiología , Regulación Neoplásica de la Expresión Génica , Ontología de Genes , Células Endoteliales de la Vena Umbilical Humana , Humanos , Técnicas In Vitro , Microscopía Intravital , Microfluídica , Microscopía Confocal , Células Neoplásicas Circulantes , Quinazolinas/farmacología , Quinazolinas/uso terapéutico , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Transducción de Señal/fisiología , Sunitinib/farmacología , Sunitinib/uso terapéutico , Migración Transendotelial y Transepitelial/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Pez Cebra/embriología
8.
EMBO Mol Med ; 13(6): e13270, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33988305

RESUMEN

Immune checkpoint therapy, where CD8 tumor infiltrating T lymphocytes (TIL) are reactivated, is a promising anti-cancer treatment approach, yet with low response rates. The extracellular matrix, in particular tenascin-C, may generate barriers for TIL. To investigate this possibility, we used a MMTV-NeuNT and syngeneic mammary gland grafting model derived thereof with engineered tenascin-C levels and observed accumulation of CD8 TIL in tenascin-C-rich stroma. Inhibition studies revealed that tenascin-C induced CXCL12 through TLR4. By binding CXCL12, tenascin-C retained CD8 TIL in the stroma. Blockade of CXCR4, the receptor of CXCL12, enhanced macrophage and CD8 TIL infiltration and reduced tumor growth and subsequent metastasis. Retention of CD8 TIL by tenascin-C/CXCL12 was also observed in human breast cancer by tissue staining. Moreover, whereas high CD8 TIL numbers correlated with longer metastasis-free survival, this was not the case when also tenascin-C and CXCL12 levels were high. Altogether, these results may be useful for improving tumor immunity as diagnostic tool and to formulate a future "TIL-matrix-release-and-reactivate" strategy.


Asunto(s)
Linfocitos Infiltrantes de Tumor , Neoplasias , Linfocitos T CD8-positivos , Quimiocina CXCL12 , Matriz Extracelular , Humanos , Tenascina
9.
Leukemia ; 35(5): 1463-1474, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33833385

RESUMEN

B-cell receptor (BCR) signaling is crucial for the pathophysiology of most mature B-cell lymphomas/leukemias and has emerged as a therapeutic target whose effectiveness remains limited by the occurrence of mutations. Therefore, deciphering the cellular program activated downstream this pathway has become of paramount importance for the development of innovative therapies. Using an original ex vivo model of BCR-induced proliferation of chronic lymphocytic leukemia cells, we generated 108 temporal transcriptional and proteomic profiles from 1 h up to 4 days after BCR activation. This dataset revealed a structured temporal response composed of 13,065 transcripts and 4027 proteins, comprising a leukemic proliferative signature consisting of 430 genes and 374 proteins. Mathematical modeling of this complex cellular response further highlighted a transcriptional network driven by 14 early genes linked to proteins involved in cell proliferation. This group includes expected genes (EGR1/2, NF-kB) and genes involved in NF-kB signaling modulation (TANK, ROHF) and immune evasion (KMO, IL4I1) that have not yet been associated with leukemic cells proliferation. Our study unveils the BCR-activated proliferative genetic program in primary leukemic cells. This approach combining temporal measurements with modeling allows identifying new putative targets for innovative therapy of lymphoid malignancies and also cancers dependent on ligand-receptor interactions.


Asunto(s)
Linfocitos B/metabolismo , Proliferación Celular/genética , Leucemia Linfocítica Crónica de Células B/genética , Receptores de Antígenos de Linfocitos B/genética , Anciano , Femenino , Humanos , Leucemia Linfocítica Crónica de Células B/metabolismo , Activación de Linfocitos/genética , Masculino , Persona de Mediana Edad , Proteoma/genética , Proteómica/métodos , Transducción de Señal/genética , Transcripción Genética/genética
10.
Elife ; 102021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-33404012

RESUMEN

Cancer extracellular vesicles (EVs) shuttle at distance and fertilize pre-metastatic niches facilitating subsequent seeding by tumor cells. However, the link between EV secretion mechanisms and their capacity to form pre-metastatic niches remains obscure. Using mouse models, we show that GTPases of the Ral family control, through the phospholipase D1, multi-vesicular bodies homeostasis and tune the biogenesis and secretion of pro-metastatic EVs. Importantly, EVs from RalA or RalB depleted cells have limited organotropic capacities in vivoand are less efficient in promoting metastasis. RalA and RalB reduce the EV levels of the adhesion molecule MCAM/CD146, which favors EV-mediated metastasis by allowing EVs targeting to the lungs. Finally, RalA, RalB, and MCAM/CD146, are factors of poor prognosis in breast cancer patients. Altogether, our study identifies RalGTPases as central molecules linking the mechanisms of EVs secretion and cargo loading to their capacity to disseminate and induce pre-metastatic niches in a CD146-dependent manner.


Asunto(s)
Neoplasias de la Mama/genética , Exosomas/patología , GTP Fosfohidrolasas/metabolismo , Metástasis de la Neoplasia/genética , Animales , Neoplasias de la Mama/secundario , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Cuerpos Multivesiculares/fisiología , Pez Cebra
11.
J Exp Med ; 217(12)2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-32766723

RESUMEN

The Nck-associated protein 1-like (NCKAP1L) gene, alternatively called hematopoietic protein 1 (HEM-1), encodes a hematopoietic lineage-specific regulator of the actin cytoskeleton. Nckap1l-deficient mice have anomalies in lymphocyte development, phagocytosis, and neutrophil migration. Here we report, for the first time, NCKAP1L deficiency cases in humans. In two unrelated patients of Middle Eastern origin, recessive mutations in NCKAP1L abolishing protein expression led to immunodeficiency, lymphoproliferation, and hyperinflammation with features of hemophagocytic lymphohistiocytosis. Immunophenotyping showed an inverted CD4/CD8 ratio with a major shift of both CD4+ and CD8+ cells toward memory compartments, in line with combined RNA-seq/proteomics analyses revealing a T cell exhaustion signature. Consistent with the core function of NCKAP1L in the reorganization of the actin cytoskeleton, patients' T cells displayed impaired early activation, immune synapse morphology, and leading edge formation. Moreover, knockdown of nckap1l in zebrafish led to defects in neutrophil migration. Hence, NCKAP1L mutations lead to broad immune dysregulation in humans, which could be classified within actinopathies.


Asunto(s)
Síndromes de Inmunodeficiencia/complicaciones , Inflamación/complicaciones , Trastornos Linfoproliferativos/complicaciones , Proteínas de la Membrana/metabolismo , Actinas/metabolismo , Animales , Degranulación de la Célula , Proliferación Celular , Niño , Citotoxicidad Inmunológica , Familia , Femenino , Homocigoto , Humanos , Síndromes de Inmunodeficiencia/inmunología , Sinapsis Inmunológicas/metabolismo , Lactante , Inflamación/inmunología , Inflamación/patología , Activación de Linfocitos/inmunología , Trastornos Linfoproliferativos/inmunología , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Mutación/genética , Linaje , Fenotipo , Síndrome , Pez Cebra
12.
Cancer Immunol Res ; 8(9): 1122-1138, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32665262

RESUMEN

Inherent immune suppression represents a major challenge in the treatment of human cancer. The extracellular matrix molecule tenascin-C promotes cancer by multiple mechanisms, yet the roles of tenascin-C in tumor immunity are incompletely understood. Using a 4NQO-induced oral squamous cell carcinoma (OSCC) model with abundant and absent tenascin-C, we demonstrated that tenascin-C enforced an immune-suppressive lymphoid stroma via CCL21/CCR7 signaling, leading to increased metastatic tumors. Through TLR4, tenascin-C increased expression of CCR7 in CD11c+ myeloid cells. By inducing CCL21 in lymphatic endothelial cells via integrin α9ß1 and binding to CCL21, tenascin-C immobilized CD11c+ cells in the stroma. Inversion of the lymph node-to-tumor CCL21 gradient, recruitment of T regulatory cells, high expression of anti-inflammatory cytokines, and matrisomal components were hallmarks of the tenascin-C-instructed lymphoid stroma. Ablation of tenascin-C or CCR7 blockade inhibited the lymphoid immune-suppressive stromal properties, reducing tumor growth, progression, and metastasis. Thus, targeting CCR7 could be relevant in human head and neck tumors, as high tenascin-C expression and an immune-suppressive stroma correlate to poor patient survival.


Asunto(s)
Neoplasias de la Boca/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Tenascina/inmunología , Animales , Quimiocina CCL21/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias de la Boca/patología , Receptores CCR7/inmunología , Proteínas Recombinantes/farmacología , Linfocitos T Reguladores/inmunología , Tenascina/farmacología , Microambiente Tumoral/inmunología
13.
Theranostics ; 10(5): 2158-2171, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32104502

RESUMEN

Rationale: The role of Monosodium Urate (MSU) crystals in gout pathophysiology is well described, as is the major impact of IL-1ß in the inflammatory reaction that constitutes the hallmark of the disease. However, despite the discovery of the NLRP3 inflammasome and its role as a Pattern Recognition Receptor linking the detection of a danger signal (MSU) to IL-1ß secretion in vitro, the precise mechanisms leading to joint inflammation in gout patients are still poorly understood. Methods: Acute urate crystal inflammation was obtained by subcutaneous injections of MSU crystals in mice. Symptoms were followed by scoring, cytokine quantification by ELISA and western blot, gene expression by RT-qPCR and RNAseq; Magnetic Resonance Imaging was also used to assess inflammation. Results: We provide an extensive clinical, biological and molecular characterization of an acute uratic inflammation mouse model which accurately mimics human gout. We report the efficacy of topical imiquimod treatment and its impact on Interferon-dependent down modulation of Il-1ß gene expression in this experimental model. Conclusion: Our work reveals several key features of MSU-dependent inflammation and identifies novel therapeutic opportunities for gout patients.


Asunto(s)
Gota/tratamiento farmacológico , Imiquimod/farmacología , Inflamación/inducido químicamente , Interleucina-1beta/efectos de los fármacos , Ácido Úrico/efectos adversos , Enfermedad Aguda , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/uso terapéutico , Administración Tópica , Animales , Antioxidantes/administración & dosificación , Antioxidantes/efectos adversos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Gota/metabolismo , Gota/patología , Imiquimod/administración & dosificación , Imiquimod/uso terapéutico , Inflamación/diagnóstico , Inflamación/inmunología , Inyecciones Subcutáneas , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Noqueados , Ácido Úrico/administración & dosificación
14.
Cancer Immunol Res ; 8(3): 368-382, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31941671

RESUMEN

The interplay between cancer cells and immune cells is a key determinant of tumor survival. Here, we uncovered how tumors exploit the immunomodulatory properties of the extracellular matrix to create a microenvironment that enables their escape from immune surveillance. Using orthotopic grafting of mammary tumor cells in immunocompetent mice and autochthonous models of breast cancer, we discovered how tenascin-C, a matrix molecule absent from most healthy adult tissues but expressed at high levels and associated with poor patient prognosis in many solid cancers, controls the immune status of the tumor microenvironment. We found that, although host-derived tenascin-C promoted immunity via recruitment of proinflammatory, antitumoral macrophages, tumor-derived tenascin-C subverted host defense by polarizing tumor-associated macrophages toward a pathogenic, immune-suppressive phenotype. Therapeutic monoclonal antibodies that blocked tenascin-C activation of Toll-like receptor 4 reversed this phenotypic switch in vitro and reduced tumor growth and lung metastasis in vivo, providing enhanced benefit in combination with anti-PD-L1 over either treatment alone. Combined tenascin-C:macrophage gene-expression signatures delineated a significant survival benefit in people with breast cancer. These data revealed a new approach to targeting tumor-specific macrophage polarization that may be effective in controlling the growth and spread of breast tumors.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Macrófagos/inmunología , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/inmunología , Femenino , Humanos , Vigilancia Inmunológica , Inmunoterapia/métodos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Ratones , Fenotipo , Tenascina/inmunología , Células Tumorales Cultivadas , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
16.
Sci Rep ; 9(1): 895, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30696890

RESUMEN

The prognosis of patients with relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL) remains unsatisfactory and, despite major advances in genomic studies, the biological mechanisms underlying chemoresistance are still poorly understood. We conducted for the first time a large-scale differential multi-omics investigation on DLBCL patient's samples in order to identify new biomarkers that could early identify patients at risk of R/R disease and to identify new targets that could determine chemorefractoriness. We compared a well-characterized cohort of R/R versus chemosensitive DLBCL patients by combining label-free quantitative proteomics and targeted RNA sequencing performed on the same tissues samples. The cross-section of both data levels allowed extracting a sub-list of 22 transcripts/proteins pairs whose expression levels significantly differed between the two groups of patients. In particular, we identified significant targets related to tumor metabolism (Hexokinase 3), microenvironment (IDO1, CXCL13), cancer cells proliferation, migration and invasion (S100 proteins) or BCR signaling pathway (CD79B). Overall, this study revealed several extremely promising biomarker candidates related to DLBCL chemorefractoriness and highlighted some new potential therapeutic drug targets. The complete datasets have been made publically available and should constitute a valuable resource for the future research.


Asunto(s)
Resistencia a Antineoplásicos/genética , Genómica , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/metabolismo , Metabolómica , Proteómica , Adolescente , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biología Computacional/métodos , Femenino , Perfilación de la Expresión Génica , Ontología de Genes , Genómica/métodos , Humanos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/patología , Masculino , Metabolómica/métodos , Persona de Mediana Edad , Estadificación de Neoplasias , Proteómica/métodos , Retratamiento , Resultado del Tratamiento , Microambiente Tumoral , Adulto Joven
17.
Am J Hum Genet ; 104(2): 319-330, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30639322

RESUMEN

ZMIZ1 is a coactivator of several transcription factors, including p53, the androgen receptor, and NOTCH1. Here, we report 19 subjects with intellectual disability and developmental delay carrying variants in ZMIZ1. The associated features include growth failure, feeding difficulties, microcephaly, facial dysmorphism, and various other congenital malformations. Of these 19, 14 unrelated subjects carried de novo heterozygous single-nucleotide variants (SNVs) or single-base insertions/deletions, 3 siblings harbored a heterozygous single-base insertion, and 2 subjects had a balanced translocation disrupting ZMIZ1 or involving a regulatory region of ZMIZ1. In total, we identified 13 point mutations that affect key protein regions, including a SUMO acceptor site, a central disordered alanine-rich motif, a proline-rich domain, and a transactivation domain. All identified variants were absent from all available exome and genome databases. In vitro, ZMIZ1 showed impaired coactivation of the androgen receptor. In vivo, overexpression of ZMIZ1 mutant alleles in developing mouse brains using in utero electroporation resulted in abnormal pyramidal neuron morphology, polarization, and positioning, underscoring the importance of ZMIZ1 in neural development and supporting mutations in ZMIZ1 as the cause of a rare neurodevelopmental syndrome.


Asunto(s)
Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Mutación Puntual , Factores de Transcripción/genética , Alelos , Animales , Niño , Preescolar , Discapacidades del Desarrollo/patología , Femenino , Humanos , Lactante , Discapacidad Intelectual/patología , Masculino , Ratones , Síndrome , Factores de Transcripción/química , Factores de Transcripción/metabolismo
18.
PLoS Pathog ; 14(10): e1007368, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30335851

RESUMEN

Infection with human BK polyomavirus, a small double-stranded DNA virus, potentially results in severe complications in immunocompromised patients. Here, we describe the in vivo variability and evolution of the BK polyomavirus by deep sequencing. Our data reveal the highest genomic evolutionary rate described in double-stranded DNA viruses, i.e., 10(-3)-10(-5) substitutions per nucleotide site per year. High mutation rates in viruses allow their escape from immune surveillance and adaptation to new hosts. By combining mutational landscapes across viral genomes with in silico prediction of viral peptides, we demonstrate the presence of significantly more coding substitutions within predicted cognate HLA-C-bound viral peptides than outside. This finding suggests a role for HLA-C in antiviral immunity, perhaps through the action of killer cell immunoglobulin-like receptors. The present study provides a comprehensive view of viral evolution and immune escape in a DNA virus.


Asunto(s)
Virus BK/genética , Antígenos HLA-C/metabolismo , Mutación , Trasplante de Órganos , Fragmentos de Péptidos/metabolismo , Infecciones por Polyomavirus/virología , Sustitución de Aminoácidos , Virus BK/inmunología , Genoma Viral , Antígenos HLA-C/genética , Antígenos HLA-C/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Filogenia , Infecciones por Polyomavirus/genética , Infecciones por Polyomavirus/inmunología
19.
Ann Rheum Dis ; 77(11): 1675-1687, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30030262

RESUMEN

OBJECTIVES: The objective of the present study was to explain why two siblings carrying both the same homozygous pathogenic mutation for the autoinflammatory disease hyper IgD syndrome, show opposite phenotypes, that is, the first being asymptomatic, the second presenting all classical characteristics of the disease. METHODS: Where single omics (mainly exome) analysis fails to identify culprit genes/mutations in human complex diseases, multiomics analyses may provide solutions, although this has been seldom used in a clinical setting. Here we combine exome, transcriptome and proteome analyses to decipher at a molecular level, the phenotypic differences between the two siblings. RESULTS: This multiomics approach led to the identification of a single gene-STAT1-which harboured a rare missense variant and showed a significant overexpression of both mRNA and protein in the symptomatic versus the asymptomatic sister. This variant was shown to be of gain of function nature, involved in an increased activation of the Janus kinase/signal transducer and activator of transcription signalling (JAK/STAT) pathway, known to play a critical role in inflammatory diseases and for which specific biotherapies presently exist. Pathway analyses based on information from differentially expressed transcripts and proteins confirmed the central role of STAT1 in the proposed regulatory network leading to an increased inflammatory phenotype in the symptomatic sibling. CONCLUSIONS: This study demonstrates the power of a multiomics approach to uncover potential clinically actionable targets for a personalised therapy. In more general terms, we provide a proteogenomics analysis pipeline that takes advantage of subject-specific genomic and transcriptomic information to improve protein identification and hence advance individualised medicine.


Asunto(s)
Genes Modificadores , Deficiencia de Mevalonato Quinasa/genética , Factor de Transcripción STAT1/genética , Adulto , Exoma , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Persona de Mediana Edad , Mutación Missense , Fenotipo , Polimorfismo de Nucleótido Simple , Proteómica/métodos
20.
Dev Cell ; 45(1): 33-52.e12, 2018 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-29634935

RESUMEN

Metastatic seeding is driven by cell-intrinsic and environmental cues, yet the contribution of biomechanics is poorly known. We aim to elucidate the impact of blood flow on the arrest and the extravasation of circulating tumor cells (CTCs) in vivo. Using the zebrafish embryo, we show that arrest of CTCs occurs in vessels with favorable flow profiles where flow forces control the adhesion efficacy of CTCs to the endothelium. We biophysically identified the threshold values of flow and adhesion forces allowing successful arrest of CTCs. In addition, flow forces fine-tune tumor cell extravasation by impairing the remodeling properties of the endothelium. Importantly, we also observe endothelial remodeling at arrest sites of CTCs in mouse brain capillaries. Finally, we observed that human supratentorial brain metastases preferably develop in areas with low perfusion. These results demonstrate that hemodynamic profiles at metastatic sites regulate key steps of extravasation preceding metastatic outgrowth.


Asunto(s)
Neoplasias Encefálicas/secundario , Neoplasias de la Mama/patología , Adhesión Celular , Hemodinámica , Neoplasias Pulmonares/patología , Melanoma/patología , Células Neoplásicas Circulantes/patología , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias de la Mama/metabolismo , Ciclo Celular , Circulación Cerebrovascular , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Melanoma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Neoplásicas Circulantes/metabolismo , Estudios Retrospectivos , Células Tumorales Cultivadas , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA