Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nat Commun ; 15(1): 3297, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740748

ABSTRACT

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.


Subject(s)
Blood Platelets , Neoplasm Metastasis , Platelet Membrane Glycoproteins , Animals , Blood Platelets/metabolism , Blood Platelets/drug effects , Humans , Mice , Platelet Membrane Glycoproteins/metabolism , Platelet Membrane Glycoproteins/genetics , Cell Line, Tumor , Female , Mice, Inbred C57BL
2.
EMBO Rep ; 24(12): e57042, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37971863

ABSTRACT

Extracellular vesicles released by tumors (tEVs) disseminate via circulatory networks and promote microenvironmental changes in distant organs favoring metastatic seeding. Despite their abundance in the bloodstream, how hemodynamics affect the function of circulating tEVs remains unsolved. We demonstrated that efficient uptake of tEVs occurs in venous endothelial cells that are subjected to hemodynamics. Low flow regimes observed in veins partially reroute internalized tEVs toward non-acidic and non-degradative Rab14-positive endosomes, at the expense of lysosomes, suggesting that endothelial mechanosensing diverts tEVs from degradation. Subsequently, tEVs promote the expression of pro-angiogenic transcription factors in low flow-stimulated endothelial cells and favor vessel sprouting in zebrafish. Altogether, we demonstrate that low flow regimes potentiate the pro-tumoral function of circulating tEVs by promoting their uptake and rerouting their trafficking. We propose that tEVs contribute to pre-metastatic niche formation by exploiting endothelial mechanosensing in specific vascular regions with permissive hemodynamics.


Subject(s)
Extracellular Vesicles , Neoplasms , Animals , Endothelial Cells , Zebrafish , Extracellular Vesicles/metabolism , Hemodynamics , Neoplasms/pathology , Angiogenesis
3.
STAR Protoc ; 4(3): 102397, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37393615

ABSTRACT

Adipose tissue plays a central role in age-related diseases. While RNAseq protocols exist for many tissues, few data have been generated with this technology to explore gene expression in adipocytes, particularly during aging. Here, we present a protocol to analyze the transcriptional changes that occur in adipose tissue during normal and accelerated aging in mouse models. We describe steps for genotyping, diet control, euthanasia, and dissection. We then detail RNA purification and genome-wide data generation and analysis. For complete details on the use and execution of this protocol, please refer to De Cauwer et al. (2022) iScience. Sep 16;25(10):105149.


Subject(s)
Adipocytes , Transcriptome , Animals , Mice , Transcriptome/genetics , Adipose Tissue , Aging/genetics , Disease Models, Animal
4.
iScience ; 25(10): 105149, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36185376

ABSTRACT

Age-related diseases are major concern in developed countries. To avoid disabilities that accompany increased lifespan, pharmaceutical approaches are considered. Therefore, appropriate animal models are required for a better understanding of aging processes and potential in vivo assays to evaluate the impact of molecules that may delay the occurrence of age-related diseases. Few mouse models exhibiting pathological aging exist, but currently, none of them reproducibly mimics human diseases like osteoporosis, cognitive dysfunctions or sarcopenia that can be seen in some, but not all, elders. Here, we describe the premature aging phenotypes of Dicer-deficient mature animals, which exhibit an overall deterioration of many organs and tissues (skin, heart, and adipose tissue) ultimately leading to a significant reduction of their lifespan. Molecular characterization of transcriptional responses focused on the adipose tissue suggested that both canonical and non-canonical functions of DICER are involved in this process and highlight potential actionable pathways to revert it.

5.
Nat Med ; 28(5): 989-998, 2022 05.
Article in English | MEDLINE | ID: mdl-35288692

ABSTRACT

The identity of histocompatibility loci, besides human leukocyte antigen (HLA), remains elusive. The major histocompatibility complex (MHC) class I MICA gene is a candidate histocompatibility locus. Here, we investigate its role in a French multicenter cohort of 1,356 kidney transplants. MICA mismatches were associated with decreased graft survival (hazard ratio (HR), 2.12; 95% confidence interval (CI): 1.45-3.11; P < 0.001). Both before and after transplantation anti-MICA donor-specific antibodies (DSA) were strongly associated with increased antibody-mediated rejection (ABMR) (HR, 3.79; 95% CI: 1.94-7.39; P < 0.001; HR, 9.92; 95% CI: 7.43-13.20; P < 0.001, respectively). This effect was synergetic with that of anti-HLA DSA before and after transplantation (HR, 25.68; 95% CI: 3.31-199.41; P = 0.002; HR, 82.67; 95% CI: 33.67-202.97; P < 0.001, respectively). De novo-developed anti-MICA DSA were the most harmful because they were also associated with reduced graft survival (HR, 1.29; 95% CI: 1.05-1.58; P = 0.014). Finally, the damaging effect of anti-MICA DSA on graft survival was confirmed in an independent cohort of 168 patients with ABMR (HR, 1.71; 95% CI: 1.02-2.86; P = 0.041). In conclusion, assessment of MICA matching and immunization for the identification of patients at high risk for transplant rejection and loss is warranted.


Subject(s)
Kidney Transplantation , Graft Rejection/genetics , Graft Survival/genetics , Histocompatibility Antigens Class I/genetics , Humans
6.
Sci Transl Med ; 14(628): eabj7521, 2022 Jan 19.
Article in English | MEDLINE | ID: mdl-34698500

ABSTRACT

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.


Subject(s)
COVID-19 , ADAM Proteins , Artificial Intelligence , Humans , Intensive Care Units , Membrane Proteins , Respiration, Artificial , SARS-CoV-2
7.
RMD Open ; 7(3)2021 10.
Article in English | MEDLINE | ID: mdl-34725261

ABSTRACT

OBJECTIVE: To explore at the molecular level the phenotype of a patient suffering an autoinflammatory syndrome which was diagnosed as familial cold autoinflammatory syndrome type 2 (FCAS-2). To explore the functions of Nlrp12 in inflammation using mouse models. METHODS: Whole exome sequencing and Nlrp12 targeted resequencing were performed on DNA isolated from the patient and her family members. In vivo and ex vivo models of inflammation (urate crystals-dependent acute joint inflammation and urate crystals-induced peritonitis) were analysed in Nlrp12-deficient and Nlrp12-competent mice. RESULTS: A rare missense NLRP12 variant (c.857C>T, p.P286L) was identified in the patient and her healthy relatives. Nlrp12-deficient mice exhibit reduced systemic inflammation and neutrophilic infiltration. CONCLUSION: Nlrp12 mediates proinflammatory functions in mice. In humans, the identification of Nlrp12 variants must be cautiously interpreted depending on clinical and paraclinical data to diagnose FCAS-2.


Subject(s)
Arthritis , Cryopyrin-Associated Periodic Syndromes , Animals , Cryopyrin-Associated Periodic Syndromes/diagnosis , Cryopyrin-Associated Periodic Syndromes/genetics , Female , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mutation, Missense , Phenotype
9.
Sci Rep ; 11(1): 13144, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162963

ABSTRACT

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.


Subject(s)
Endothelium, Vascular/physiology , Hemorheology , Transendothelial and Transepithelial Migration , Animals , Animals, Genetically Modified , Blood Flow Velocity/drug effects , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/physiology , Gene Expression Regulation, Neoplastic , Gene Ontology , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Intravital Microscopy , Microfluidics , Microscopy, Confocal , Neoplastic Cells, Circulating , Quinazolines/pharmacology , Quinazolines/therapeutic use , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Signal Transduction/physiology , Sunitinib/pharmacology , Sunitinib/therapeutic use , Transendothelial and Transepithelial Migration/drug effects , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/physiology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/physiology , Zebrafish/embryology
10.
Leukemia ; 35(5): 1463-1474, 2021 05.
Article in English | MEDLINE | ID: mdl-33833385

ABSTRACT

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.


Subject(s)
B-Lymphocytes/metabolism , Cell Proliferation/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Receptors, Antigen, B-Cell/genetics , Aged , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymphocyte Activation/genetics , Male , Middle Aged , Proteome/genetics , Proteomics/methods , Signal Transduction/genetics , Transcription, Genetic/genetics
11.
J Exp Med ; 217(12)2020 12 07.
Article in English | MEDLINE | ID: mdl-32766723

ABSTRACT

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.


Subject(s)
Immunologic Deficiency Syndromes/complications , Inflammation/complications , Lymphoproliferative Disorders/complications , Membrane Proteins/metabolism , Actins/metabolism , Animals , Cell Degranulation , Cell Proliferation , Child , Cytotoxicity, Immunologic , Family , Female , Homozygote , Humans , Immunologic Deficiency Syndromes/immunology , Immunological Synapses/metabolism , Infant , Inflammation/immunology , Inflammation/pathology , Lymphocyte Activation/immunology , Lymphoproliferative Disorders/immunology , Male , Membrane Proteins/chemistry , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mutation/genetics , Pedigree , Phenotype , Syndrome , Zebrafish
12.
Bone Marrow Transplant ; 55(7): 1367-1378, 2020 07.
Article in English | MEDLINE | ID: mdl-32286503

ABSTRACT

Graft-versus-host disease (GVHD) and cytomegalovirus (CMV)-related complications are leading causes of mortality after unrelated-donor hematopoietic cell transplantation (UD-HCT). The non-conventional MHC class I gene MICB, alike MICA, encodes a stress-induced polymorphic NKG2D ligand. However, unlike MICA, MICB interacts with the CMV-encoded UL16, which sequestrates MICB intracellularly, leading to immune evasion. Here, we retrospectively analyzed the impact of mismatches in MICB amino acid position 98 (MICB98), a key polymorphic residue involved in UL16 binding, in 943 UD-HCT pairs who were allele-matched at HLA-A, -B, -C, -DRB1, -DQB1 and MICA loci. HLA-DP typing was further available. MICB98 mismatches were significantly associated with an increased incidence of acute (grade II-IV: HR, 1.20; 95% CI, 1.15 to 1.24; P < 0.001; grade III-IV: HR, 2.28; 95% CI, 1.56 to 3.34; P < 0.001) and chronic GVHD (HR, 1.21; 95% CI, 1.10 to 1.33; P < 0.001). MICB98 matching significantly reduced the effect of CMV status on overall mortality from a hazard ratio of 1.77 to 1.16. MICB98 mismatches showed a GVHD-independent association with a higher incidence of CMV infection/reactivation (HR, 1.84; 95% CI, 1.34 to 2.51; P < 0.001). Hence selecting a MICB98-matched donor significantly reduces the GVHD incidence and lowers the impact of CMV status on overall survival.


Subject(s)
Cytomegalovirus Infections , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Amino Acids , Cytomegalovirus Infections/epidemiology , Cytomegalovirus Infections/prevention & control , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Incidence , Retrospective Studies
13.
Theranostics ; 10(5): 2158-2171, 2020.
Article in English | MEDLINE | ID: mdl-32104502

ABSTRACT

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.


Subject(s)
Gout/drug therapy , Imiquimod/pharmacology , Inflammation/chemically induced , Interleukin-1beta/drug effects , Uric Acid/adverse effects , Acute Disease , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Administration, Topical , Animals , Antioxidants/administration & dosage , Antioxidants/adverse effects , Cytokines/metabolism , Disease Models, Animal , Gout/metabolism , Gout/pathology , Imiquimod/administration & dosage , Imiquimod/therapeutic use , Inflammation/diagnosis , Inflammation/immunology , Injections, Subcutaneous , Magnetic Resonance Imaging/methods , Mice , Mice, Knockout , Uric Acid/administration & dosage
15.
J Hum Genet ; 64(7): 689-694, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31068678

ABSTRACT

Cathepsin C (CatC) is a cysteine protease involved in a variety of immune and inflammatory pathways such as activation of cytotoxicity of various immune cells. Homozygous or compound heterozygous variants in the CatC coding gene CTSC cause different conditions that have in common severe periodontitis. Periodontitis may occur as part of Papillon-Lefèvre syndrome (PLS; OMIM#245000) or Haim-Munk syndrome (HMS; OMIM#245010), or may present as an isolated finding named aggressive periodontitis (AP1; OMIM#170650). AP1 generally affects young children and results in destruction of the periodontal support of the primary dentition. In the present study we report exome sequencing of a three generation consanguineous Turkish family with a recessive form of early-onset AP1. We identified a novel homozygous missense variant in exon 2 of CTSC (NM_148170, c.G302C, p.Trp101Ser) predicted to disrupt protein structure and to be disease causing. This is the first described CTSC variant specific to the nonsyndromic AP1 form. Given the broad phenotypic spectrum associated with CTSC variants, reporting this novel variant gives new insights on genotype/phenotype correlations and might improve diagnosis of patients with early-onset AP1.


Subject(s)
Aggressive Periodontitis/genetics , Cathepsin C/genetics , Mutation, Missense , Adolescent , Adult , Aggressive Periodontitis/diagnostic imaging , Cathepsin C/chemistry , Child , Female , Genetic Association Studies , Homozygote , Humans , Male , Turkey , Exome Sequencing
16.
Sci Rep ; 9(1): 895, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30696890

ABSTRACT

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.


Subject(s)
Drug Resistance, Neoplasm/genetics , Genomics , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Metabolomics , Proteomics , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Computational Biology/methods , Female , Gene Expression Profiling , Gene Ontology , Genomics/methods , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Metabolomics/methods , Middle Aged , Neoplasm Staging , Proteomics/methods , Retreatment , Treatment Outcome , Tumor Microenvironment , Young Adult
17.
Am J Hum Genet ; 104(2): 319-330, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30639322

ABSTRACT

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.


Subject(s)
Developmental Disabilities/genetics , Intellectual Disability/genetics , Point Mutation , Transcription Factors/genetics , Alleles , Animals , Child , Child, Preschool , Developmental Disabilities/pathology , Female , Humans , Infant , Intellectual Disability/pathology , Male , Mice , Syndrome , Transcription Factors/chemistry , Transcription Factors/metabolism
18.
Ann Rheum Dis ; 77(11): 1675-1687, 2018 11.
Article in English | MEDLINE | ID: mdl-30030262

ABSTRACT

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.


Subject(s)
Genes, Modifier , Mevalonate Kinase Deficiency/genetics , STAT1 Transcription Factor/genetics , Adult , Exome , Female , Gene Expression Profiling/methods , Humans , Middle Aged , Mutation, Missense , Phenotype , Polymorphism, Single Nucleotide , Proteomics/methods
19.
Dev Cell ; 45(1): 33-52.e12, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29634935

ABSTRACT

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.


Subject(s)
Brain Neoplasms/secondary , Breast Neoplasms/pathology , Cell Adhesion , Hemodynamics , Lung Neoplasms/pathology , Melanoma/pathology , Neoplastic Cells, Circulating/pathology , Animals , Brain Neoplasms/metabolism , Breast Neoplasms/metabolism , Cell Cycle , Cerebrovascular Circulation , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Humans , Lung Neoplasms/metabolism , Male , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Cells, Circulating/metabolism , Retrospective Studies , Tumor Cells, Cultured , Zebrafish
20.
J Clin Invest ; 127(11): 4090-4103, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28972538

ABSTRACT

Shwachman-Diamond syndrome (SDS) (OMIM #260400) is a rare inherited bone marrow failure syndrome (IBMFS) that is primarily characterized by neutropenia and exocrine pancreatic insufficiency. Seventy-five to ninety percent of patients have compound heterozygous loss-of-function mutations in the Shwachman-Bodian-Diamond syndrome (sbds) gene. Using trio whole-exome sequencing (WES) in an sbds-negative SDS family and candidate gene sequencing in additional SBDS-negative SDS cases or molecularly undiagnosed IBMFS cases, we identified 3 independent patients, each of whom carried a de novo missense variant in srp54 (encoding signal recognition particle 54 kDa). These 3 patients shared congenital neutropenia linked with various other SDS phenotypes. 3D protein modeling revealed that the 3 variants affect highly conserved amino acids within the GTPase domain of the protein that are critical for GTP and receptor binding. Indeed, we observed that the GTPase activity of the mutated proteins was impaired. The level of SRP54 mRNA in the bone marrow was 3.6-fold lower in patients with SRP54-mutations than in healthy controls. Profound reductions in neutrophil counts and chemotaxis as well as a diminished exocrine pancreas size in a SRP54-knockdown zebrafish model faithfully recapitulated the human phenotype. In conclusion, autosomal dominant mutations in SRP54, a key member of the cotranslation protein-targeting pathway, lead to syndromic neutropenia with a Shwachman-Diamond-like phenotype.


Subject(s)
Bone Marrow Diseases/genetics , Exocrine Pancreatic Insufficiency/genetics , Lipomatosis/genetics , Neutropenia/congenital , Signal Recognition Particle/genetics , Animals , Child , Congenital Bone Marrow Failure Syndromes , DNA Mutational Analysis , Female , Genetic Association Studies , Humans , Infant , Male , Models, Molecular , Neutropenia/genetics , Pancreas, Exocrine/metabolism , Phenotype , Protein Domains , Shwachman-Diamond Syndrome , Signal Recognition Particle/chemistry , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...