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










Publication year range
1.
APL Bioeng ; 6(3): 036101, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35818479

ABSTRACT

In many leukemia patients, a poor prognosis is attributed either to the development of chemotherapy resistance by leukemic stem cells (LSCs) or to the inefficient engraftment of transplanted hematopoietic stem/progenitor cells (HSPCs) into the bone marrow (BM). Here, we build a 3D in vitro model system of bone marrow organoids (BMOs) that recapitulate several structural and cellular components of native BM. These organoids are formed in a high-throughput manner from the aggregation of endothelial and mesenchymal cells within hydrogel microwells. Accordingly, the mesenchymal compartment shows partial maintenance of its self-renewal and multilineage potential, while endothelial cells self-organize into an interconnected vessel-like network. Intriguingly, such an endothelial compartment enhances the recruitment of HSPCs in a chemokine ligand/receptor-dependent manner, reminiscent of HSPC homing behavior in vivo. Additionally, we also model LSC migration and nesting in BMOs, thus highlighting the potential of this system as a well accessible and scalable preclinical model for candidate drug screening and patient-specific assays.

2.
Int J Mol Sci ; 23(9)2022 May 05.
Article in English | MEDLINE | ID: mdl-35563552

ABSTRACT

Infantile hemangioma (IH) is the most common infantile tumor, affecting 5-10% of newborns. Propranolol, a nonselective ß-adrenergic receptor (ADRB) antagonist, is currently the first-line treatment for severe IH; however, both its mechanism of action and its main cellular target remain poorly understood. Since betablockers can antagonize the effect of natural ADRB agonists, we postulated that the catecholamine produced in situ in IH may have a role in the propranolol response. By quantifying catecholamines in the IH tissues, we found a higher amount of noradrenaline (NA) in untreated proliferative IHs than in involuted IHs or propranolol-treated IHs. We further found that the first three enzymes of the catecholamine biosynthesis pathway are expressed by IH cells and that their levels are reduced in propranolol-treated tumors. To study the role of NA in the pathophysiology of IH and its response to propranolol, we performed an in vitro angiogenesis assay in which IH-derived endothelial cells, pericytes and/or telocytes were incorporated. The results showed that the total tube formation is sensitive to propranolol only when exogenous NA is added in the three-cell model. We conclude that the IH's sensitivity to propranolol depends on crosstalk between the endothelial cells, pericytes and telocytes in the context of a high local amount of local NA.


Subject(s)
Hemangioma , Neuroendocrine Tumors , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Endothelial Cells/metabolism , Hemangioma/drug therapy , Hemangioma/pathology , Humans , Infant , Infant, Newborn , Neuroendocrine Tumors/metabolism , Norepinephrine/metabolism , Propranolol/pharmacology , Propranolol/therapeutic use
3.
Cancers (Basel) ; 13(18)2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34572851

ABSTRACT

The role of the proangiogenic factor olfactomedin-like 3 (OLFML3) in cancer is unclear. To characterize OLFML3 expression in human cancer and its role during tumor development, we undertook tissue expression studies, gene expression analyses of patient tumor samples, in vivo studies in mouse cancer models, and in vitro coculture experiments. OLFML3 was expressed at high levels, mainly in blood vessels, in multiple human cancers. We focused on colorectal cancer (CRC), as elevated expression of OLFML3 mRNA correlated with shorter relapse-free survival, higher tumor grade, and angiogenic microsatellite stable consensus molecular subtype 4 (CMS4). Treatment of multiple in vivo tumor models with OLFML3-blocking antibodies and deletion of the Olfml3 gene from mice decreased lymphangiogenesis, pericyte coverage, and tumor growth. Antibody-mediated blockade of OLFML3 and deletion of host Olfml3 decreased the recruitment of tumor-promoting tumor-associated macrophages and increased infiltration of the tumor microenvironment by NKT cells. Importantly, targeting OLFML3 increased the antitumor efficacy of anti-PD-1 checkpoint inhibitor therapy. Taken together, the results demonstrate that OLFML3 is a promising candidate therapeutic target for CRC.

4.
Cancers (Basel) ; 14(1)2021 Dec 23.
Article in English | MEDLINE | ID: mdl-35008226

ABSTRACT

Extracellular vesicles (EVs) act in cell-to-cell communication, delivering cargo from donor to recipient cells and modulating their physiological condition. EVs secreted by leukemic blasts in patients with leukemia have been shown to influence the fate of recipient cells in the bone marrow microenvironment. Methods to quantify and to characterize them phenotypically are therefore urgently needed to study their functional role in leukemia development and to evaluate their potential as targets for therapy. We have used cryo-electron microscopy to study morphology and size of leukemic EVs, and nanoparticle tracking analysis and fluorescence triggering flow cytometry to quantify EVs in platelet-free plasma from a small cohort of leukemia patients and healthy blood donors. Additional studies with a capture bead-based assay allowed us to establish phenotypic signatures of leukemic EVs from 17 AML and 3 B-ALL patients by evaluating the expression of 37 surface antigens. In addition to tetraspanins and lineage-specific markers we found several adhesion molecules (CD29, and CD146) to be highly expressed by EVs from B-ALL and several leukemic stem cell antigens (CD44, CD105, CD133, and SSEA-4) to be expressed by EVs from AML patients. Further improvements in analytical methods to study EVs are needed before potentially using them as biomarkers for leukemia prognosis and follow-up.

5.
FASEB J ; 34(11): 15559-15576, 2020 11.
Article in English | MEDLINE | ID: mdl-32997357

ABSTRACT

Pericytes promote vessel stability and their dysfunction causes pathologies due to blood vessel leakage. Previously, we reported that Olfactomedin-like 3 (Olfml3) is a matricellular protein with proangiogenic properties. Here, we explored the role of Olfml3 in a knockout mouse model engineered to suppress this protein. The mutant mice exhibited vascular defects in pericyte coverage, suggesting that pericytes influence blood vessel formation in an Olfml3-dependent manner. Olfml3-deficient mice exhibited abnormalities in the vasculature causing partial lethality of embryos and neonates. Reduced pericyte coverage was observed at embryonic day 12.5 and persisted throughout development, resulting in perinatal death of 35% of Olfml3-deficient mice. Cultured Olfml3-deficient pericytes exhibited aberrant motility and altered pericyte association to endothelial cells. Furthermore, the proliferative response of Olfml3-/- pericytes upon PDGF-B stimulation was significantly diminished. Subsequent experiments revealed that intact PDGF-B signaling, mediated via Olfml3 binding, is required for pericyte proliferation and activation of downstream kinase pathways. Our findings suggest a model wherein pericyte recruitment to endothelial cells requires Olfml3 to provide early instructive cue and retain PDGF-B along newly formed vessels to achieve optimal angiogenesis.


Subject(s)
Cell Movement , Cell Proliferation , Glycoproteins/physiology , Neovascularization, Pathologic/pathology , Pericytes/pathology , Proto-Oncogene Proteins c-sis/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Embryonic Development , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/metabolism , Pericytes/metabolism , Pregnancy , Signal Transduction
6.
Cancers (Basel) ; 11(10)2019 Oct 22.
Article in English | MEDLINE | ID: mdl-31652588

ABSTRACT

A major limitation of clinically used cancer drugs is the lack of specificity resulting in toxicity. To address this, we performed a phenotypically-driven screen to identify optimal multidrug combinations acting with high efficacy and selectivity in clear cell renal cell carcinoma (ccRCC). The search was performed using the Therapeutically Guided Multidrug Optimization (TGMO) method in ccRCC cells (786-O) and nonmalignant renal cells and identified a synergistic low-dose four-drug combination (C2) with high efficacy and negligible toxicity. We discovered that C2 inhibits multipolar spindle pole clustering, a survival mechanism employed by cancer cells with spindle abnormalities. This phenotype was also observed in 786-O cells resistant to sunitinib, the first line ccRCC treatment, as well as in melanoma cells with distinct percentages of supernumerary centrosomes. We conclude that C2-treatment shows a high efficacy in cells prone to form multipolar spindles. Our data suggest a highly effective and selective C2 treatment strategy for malignant and drug-resistant cancers.

7.
Br J Cancer ; 121(2): 139-149, 2019 07.
Article in English | MEDLINE | ID: mdl-31235865

ABSTRACT

BACKGROUND: Crenolanib is a tyrosine kinase inhibitor targeting PDGFR-α, PDGFR-ß and Fms related tyrosine kinase-3 (FLT3) that is currently evaluated in several clinical trials. Although platelet-derived growth factor receptor (PDGFR) signalling pathway is believed to play an important role in angiogenesis and maintenance of functional vasculature, we here demonstrate a direct angiostatic activity of crenolanib independently of PDGFR signalling. METHODS: The activity of crenolanib on cell viability, migration, sprouting, apoptosis and mitosis was assessed in endothelial cells, tumour cells and fibroblasts. Alterations in cell morphology were determined by immunofluorescence experiments. Flow-cytometry analysis and mRNA expression profiles were used to investigate cell differentiation. In vivo efficacy was investigated in human ovarian carcinoma implanted on the chicken chorioallantoic membrane (CAM). RESULTS: Crenolanib was found to inhibit endothelial cell viability, migration and sprout length, and induced apoptosis independently of PDGFR expression. Treated cells  showed altered actin arrangement and nuclear aberrations. Mitosis was affected at several levels including mitosis entry and centrosome clustering. Crenolanib suppressed human ovarian carcinoma tumour growth and angiogenesis in the CAM model. CONCLUSIONS: The PDGFR/FLT3 inhibitor crenolanib targets angiogenesis and inhibits tumour growth in vivo unrelated to PDGFR expression. Based on our findings, we suggest a broad mechanism of action of crenolanib.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Mitosis Modulators/pharmacology , Piperidines/pharmacology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Chickens , Female , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Receptors, Platelet-Derived Growth Factor/analysis , Receptors, Platelet-Derived Growth Factor/physiology
8.
Wound Repair Regen ; 24(6): 1030-1035, 2016 11.
Article in English | MEDLINE | ID: mdl-27684720

ABSTRACT

Three-dimensional biomimetic scaffolds resembling the native extracellular matrix (ECM) are widely used in tissue engineering, however they often lack optimal bioactive cues needed for acceleration of cell proliferation, neovascularization, and tissue regeneration. In this study, the use of the ECM-related protein Olfactomedin-like 3 (Olfml3) demonstrates the importance and feasibility of fabricating efficient bioactive scaffolds without in vitro cell seeding prior to in vivo implantation. First, in vivo proangiogenic properties of Olfml3 were shown in a murine wound healing model by accelerated wound closure and a 1.4-fold increase in wound vascularity. Second, subcutaneous implantation of tubular scaffolds coated with recombinant Olfml3 resulted in enhanced cell in-growth and neovascularization compared with control scaffolds. Together, our data indicates the potential of Olfml3 to accelerate neovascularization during tissue regeneration by promoting endothelial cell proliferation and migration. This study provides a promising concept for the reconstruction of damaged tissue using affordable and effective bioactive scaffolds.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biomimetic Materials , Extracellular Matrix Proteins/pharmacology , Extracellular Matrix/metabolism , Glycoproteins/pharmacology , Regeneration , Tissue Scaffolds , Wound Healing/drug effects , Wounds and Injuries/pathology , Animals , Biomimetic Materials/pharmacology , Disease Models, Animal , Female , Mice , Regenerative Medicine , Tensile Strength , Tissue Engineering/methods
9.
PLoS One ; 11(7): e0159679, 2016.
Article in English | MEDLINE | ID: mdl-27442505

ABSTRACT

Atherosclerosis, caused in part by monocytes in plaques, continues to be a disease that afflicts the modern world. Whilst significant steps have been made in treating this chronic inflammatory disease, questions remain on how to prevent monocyte and macrophage accumulation in atherosclerotic plaques. Junctional Adhesion Molecule C (JAM-C) expressed by vascular endothelium directs monocyte transendothelial migration in a unidirectional manner leading to increased inflammation. Here we show that interfering with JAM-C allows reverse-transendothelial migration of monocyte-derived cells, opening the way back out of the inflamed environment. To study the role of JAM-C in plaque regression we used a mouse model of atherosclerosis, and tested the impact of vascular JAM-C expression levels on monocyte reverse transendothelial migration using human cells. Studies in-vitro under inflammatory conditions revealed that overexpression or gene silencing of JAM-C in human endothelium exposed to flow resulted in higher rates of monocyte reverse-transendothelial migration, similar to antibody blockade. We then transplanted atherosclerotic, plaque-containing aortic arches from hyperlipidemic ApoE-/- mice into wild-type normolipidemic recipient mice. JAM-C blockade in the recipients induced greater emigration of monocyte-derived cells and further diminished the size of atherosclerotic plaques. Our findings have shown that JAM-C forms a one-way vascular barrier for leukocyte transendothelial migration only when present at homeostatic copy numbers. We have also shown that blocking JAM-C can reduce the number of atherogenic monocytes/macrophages in plaques by emigration, providing a novel therapeutic strategy for chronic inflammatory pathologies.


Subject(s)
Cell Movement , Junctional Adhesion Molecule C/metabolism , Monocytes/pathology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Animals , Antibodies/pharmacology , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Carotid Arteries/pathology , Cell Movement/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Flow Cytometry , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/pathology , Intercellular Junctions/drug effects , Intercellular Junctions/metabolism , Leukocytes/drug effects , Leukocytes/pathology , Lipids/blood , Mice, Inbred C57BL , Models, Biological , Monocytes/drug effects , Plaque, Atherosclerotic/blood
10.
FASEB J ; 29(8): 3411-25, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25911611

ABSTRACT

De novo formation of blood vessels is a pivotal mechanism during cancer development. During the past few years, antiangiogenic drugs have been developed to target tumor vasculature. However, because of limitations and adverse effects observed with current therapies, there is a strong need for alternative antiangiogenic strategies. Using specific anti-junctional adhesion molecule (JAM)-B antibodies and Jam-b-deficient mice, we studied the role in antiangiogenesis of JAM-B. We found that antibodies against murine JAM-B, an endothelium-specific adhesion molecule, inhibited microvessel outgrowth from ex vivo aortic rings and in vitro endothelial network formation. In addition, anti-JAM-B antibodies blocked VEGF signaling, an essential pathway for angiogenesis. Moreover, increased aortic ring branching was observed in aortas isolated from Jam-b-deficient animals, suggesting that JAM-B negatively regulates proangiogenic pathways. In mice, JAM-B expression was detected in de novo-formed blood vessels of tumors, but anti-JAM-B antibodies unexpectedly did not reduce tumor growth. Accordingly, JAM-B deficiency in vivo had no impact on blood vessel formation, suggesting that targeting JAM-B in vivo may be offset by other proangiogenic mechanisms. In conclusion, despite the promising effects observed in vitro, targeting JAM-B during tumor progression seems to be inefficient as a stand-alone antiangiogenesis therapy.


Subject(s)
Cell Adhesion Molecules/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Line, Tumor , Female , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
11.
Mol Cancer Ther ; 11(12): 2588-99, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23002094

ABSTRACT

Antiangiogenic drugs have been used as anticancer agents to target tumor endothelial cells or pericytes. Because of limited efficacy of the current monotherapies, there is a strong demand for the dual targeting of endothelial cells and pericytes. Here, we identify Olfactomedin-like 3 (Olfml3) as a novel proangiogenic cue within the tumor microenvironment. Tumor-derived Olfml3 is produced by both tumor endothelial cells and accompanying pericytes and deposited in the perivascular compartment. Blockade of Olfml3 by anti-Olfml3 antibodies is highly effective in reducing tumor vascularization, pericyte coverage, and tumor growth. In vitro, Olfml3 targeting is sufficient to inhibit endothelioma cell migration and sprouting. Olfml3 alone or through binding to BMP4 enhances the canonical SMAD1/5/8 signaling pathway required for BMP4-induced angiogenesis. Therefore, Olfml3 blockade provides a novel strategy to control tumor growth by targeting two distinct cell types within the tumor microenvironment using a single molecule.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/drug therapy , Glycoproteins/antagonists & inhibitors , Pericytes/drug effects , Pericytes/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Bone Morphogenetic Protein 4/metabolism , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Female , Gene Silencing , Glycoproteins/biosynthesis , Glycoproteins/genetics , Glycoproteins/immunology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Pericytes/pathology , Signal Transduction , Smad Proteins/metabolism , Swine , Transfection
12.
FASEB J ; 23(12): 4105-16, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19667118

ABSTRACT

On appropriate stimuli, quiescent endothelial cells start to proliferate and form de novo blood vessels through angiogenesis. To further define molecular mechanisms accompanying the activation of endothelial cells during angiogenesis, we identified genes that were differentially regulated during this process using microarray analyses. In this work, we established a regulatory role for Sushi repeat protein X-linked 2 (Srpx2) in endothelial cell remodeling during angiogenesis. In particular, silencing of Srpx2 using small interfering RNAs (siRNAs) specifically attenuated endothelial cell migration and delayed angiogenic sprout formation. In vivo, Srpx2 expression was detected in de novo formation of blood vessels in angiogenic tissues by in situ mRNA hybridization and immunostaining. Pulldown experiments identified Srpx2 as a ligand for vascular uPAR, a key molecule involved in invasive migration of angiogenic endothelium. Immunostaining revealed coexpression of the Srpx2 and uPAR on vascular endothelium. These findings suggest that Srpx2 regulates endothelial cell migration and tube formation and provides a new target for modulating angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Profiling , Neovascularization, Physiologic/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Gene Expression Regulation/physiology , Gene Silencing , Humans , Membrane Proteins , Neoplasm Proteins , Protein Array Analysis , Receptors, Urokinase Plasminogen Activator/metabolism
13.
Dev Biol ; 332(1): 2-24, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19465018

ABSTRACT

New perspectives on the origin of neurogenesis emerged with the identification of genes encoding post-synaptic proteins as well as many "neurogenic" regulators as the NK, Six, Pax, bHLH proteins in the Demosponge genome, a species that might differentiate sensory cells but no neurons. However, poriferans seem to miss some key regulators of the neurogenic circuitry as the Hox/paraHox and Otx-like gene families. Moreover as a general feature, many gene families encoding evolutionarily-conserved signaling proteins and transcription factors were submitted to a wave of gene duplication in the last common eumetazoan ancestor, after Porifera divergence. In contrast gene duplications in the last common bilaterian ancestor, Urbilateria, are limited, except for the bHLH Atonal-class. Hence Cnidaria share with Bilateria a large number of genetic tools. The expression and functional analyses currently available suggest a neurogenic function for numerous orthologs in developing or adult cnidarians where neurogenesis takes place continuously. As an example, in the Hydra polyp, the Clytia medusa and the Acropora coral, the Gsx/cnox2/Anthox-2 ParaHox gene likely supports neurogenesis. Also neurons and nematocytes (mechanosensory cells) share in hydrozoans a common stem cell and several regulatory genes indicating that they can be considered as sister cells. Performed in anthozoan and medusozoan species, these studies should tell us more about the way(s) evolution hazards achieved the transition from epithelial to neuronal cell fate, and about the robustness of the genetic circuitry that allowed neuromuscular transmission to arise and be maintained across evolution.


Subject(s)
Biological Evolution , Cnidaria/growth & development , Cnidaria/genetics , Neurogenesis , Animals , Cnidaria/anatomy & histology , Nervous System/anatomy & histology , Nervous System/growth & development , Neurogenesis/genetics
14.
Blood ; 110(7): 2545-55, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17625065

ABSTRACT

Monocyte recruitment from the vasculature involves sequential engagement of multiple receptors, culminating in transendothelial migration and extravasation. Junctional adhesion molecule-C (JAM-C) is localized at endothelial intercellular junctions and plays a role in monocyte transmigration. Here, we show that blockade of JAM-B/-C interaction reduced monocyte numbers in the extravascular compartment through increased reverse transmigration rather than by reduced transmigration. This was confirmed in vivo, showing that an anti-JAM-C antibody reduced the number of monocytes in inflammatory tissue and increased the number of monocytes with a reverse-transmigratory phenotype in the peripheral blood. All together, our results suggest a novel mechanism of reducing accumulation of monocytes at inflammation sites by disruption of JAM-C-mediated monocyte retention.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Movement , Monocytes/cytology , Monocytes/metabolism , Animals , Antibodies/immunology , Blood Platelets/metabolism , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cells, Cultured , Endothelial Cells/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Junctional Adhesion Molecules , Mice , Mice, Transgenic , Phenotype
15.
C R Biol ; 330(6-7): 491-7, 2007.
Article in English | MEDLINE | ID: mdl-17631443

ABSTRACT

The recent establishment of gene silencing through RNA interference upon feeding opens avenues to decipher the genetic control of regeneration in hydra. Following that approach, we identified three main stages for head regeneration. Immediately post-amputation, the serine protease inhibitor Kazal1 gene produced by the gland cells prevents from an excessive autophagy in regenerating tips. This cytoprotective function, or self-preservation, is similar to that played by Kazal-type proteins in the mammalian exocrine pancreas, in homeostatic or post-injury conditions, likely reflecting an evolutionarily conserved mechanism linking cell survival to tissue repair. Indeed, in wild-type hydra, within the first hours following mid-gastric section, an extensive cellular remodelling is taking place, including phenotypic cellular transitions and cell proliferation. The activation of the MAPK pathway, which leads to the RSK-dependent CREB phosphorylation, is required for these early cellular events. Later, at the early-late stage, the expression of the Gsx/cnox-2 ParaHox gene in proliferating apical neuronal progenitors is required for the de novo neurogenesis that precedes the emergence of the tentacle rudiments. Hence, head regeneration in wild-type hydra relies on spatially restricted and timely orchestrated cellular modifications, which display similarities with those reported during vertebrate epimorphic regeneration. These results suggest some conservation across evolution of the mechanisms driving the post-amputation reactivation of developmental programs.


Subject(s)
Hydra/cytology , Hydra/growth & development , RNA Interference/physiology , Regeneration/genetics , Regeneration/physiology , Animals , Biological Evolution , Cyclic AMP Response Element-Binding Protein/metabolism , Glycogen Synthase Kinase 3/metabolism , Nervous System/cytology , Nervous System/growth & development , Neuronal Plasticity/physiology , Phenotype , Phosphorylation , Protein Kinases/metabolism , Ribosomal Protein S6 Kinases , Serine Proteinase Inhibitors/pharmacology
16.
Development ; 134(6): 1191-201, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17301084

ABSTRACT

Because head regeneration occurs in nerve-free hydra mutants, neurogenesis was regarded as dispensable for this process. Here, in wild-type hydra, we tested the function of the ParaHox gsx homolog gene, cnox-2, which is a specific marker for bipotent neuronal progenitors, expressed in cycling interstitial cells that give rise to apical neurons and gastric nematoblasts (i.e. sensory mechanoreceptor precursors). cnox-2 RNAi silencing leads to a dramatic downregulation of hyZic, prdl-a, gsc and cnASH, whereas hyCOUP-TF is upregulated. cnox-2 indeed acts as an upstream regulator of the neuronal and nematocyte differentiation pathways, as cnox-2(-) hydra display a drastic reduction in apical neurons and gastric nematoblasts, a disorganized apical nervous system and a decreased body size. During head regeneration, the locally restricted de novo neurogenesis that precedes head formation is cnox-2 dependent: cnox-2 expression is induced in neuronal precursors and differentiating neurons that appear in the regenerating tip; cnox-2 RNAi silencing reduces this de novo neurogenesis and delays head formation. Similarly, the disappearance of cnox-2(+) cells in sf-1 mutants also correlates with head regeneration blockade. Hence in wild-type hydra, head regeneration requires the cnox-2 neurogenic function. When neurogenesis is missing, an alternative, slower and less efficient, head developmental program is possibly activated.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Hydra/physiology , Nervous System/growth & development , Regeneration/genetics , Animals , Body Patterning/genetics , Cell Differentiation/genetics , Cell Proliferation , Down-Regulation , Head/physiology , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Hydra/genetics , Nervous System/cytology , RNA Interference
17.
Semin Cell Dev Biol ; 17(4): 492-502, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16807002

ABSTRACT

The silencing of genes whose expression is restricted to specific cell types and/or specific regeneration stages opens avenues to decipher the molecular control of the cellular plasticity underlying head regeneration in hydra. In this review, we highlight recent studies that identified genes involved in the immediate cytoprotective function played by gland cells after amputation; the early dedifferentiation of digestive cells into blastema-like cells during head regeneration, and the early late proliferation of neuronal progenitors required for head patterning. Hence, developmental plasticity in hydra relies on spatially restricted and timely orchestrated cellular modifications, where the functions played by stem cells remain to be characterized.


Subject(s)
Hydra/cytology , Hydra/physiology , Multipotent Stem Cells/cytology , Animals , Autophagy/physiology , Cytoprotection/physiology , Homeostasis/physiology , Hydra/growth & development , Models, Biological , Phylogeny , Regeneration/physiology , Survival/physiology
18.
J Cell Sci ; 119(Pt 5): 846-57, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16478786

ABSTRACT

In hydra, the endodermal epithelial cells carry out the digestive function together with the gland cells that produce zymogens and express the evolutionarily conserved gene Kazal1. To assess the hydra Kazal1 function, we silenced gene expression through double-stranded RNA feeding. A progressive Kazal1 silencing affected homeostatic conditions as evidenced by the low budding rate and the induced animal death. Concomitantly, a dramatic disorganization followed by a massive death of gland cells was observed, whereas the cytoplasm of digestive cells became highly vacuolated. The presence of mitochondria and late endosomes within those vacuoles assigned them as autophagosomes. The enhanced Kazal1 expression in regenerating tips was strongly diminished in Kazal1(-) hydra, and the amputation stress led to an immediate disorganization of the gland cells, vacuolization of the digestive cells and death after prolonged silencing. This first cellular phenotype resulting from a gene knock-down in cnidarians suggests that the Kazal1 serine-protease-inhibitor activity is required to prevent excessive autophagy in intact hydra and to exert a cytoprotective function to survive the amputation stress. Interestingly, these functions parallel the pancreatic autophagy phenotype observed upon mutation within the Kazal domain of the SPINK1 and SPINK3 genes in human and mice, respectively.


Subject(s)
Carrier Proteins/genetics , Gene Silencing , Hydra/enzymology , Pancreas/metabolism , Serine Proteinase Inhibitors/genetics , Amino Acid Sequence , Animals , Gene Expression Regulation , Humans , Hydra/cytology , Hydra/genetics , Molecular Mimicry , Molecular Sequence Data , Phenotype , RNA, Double-Stranded/genetics , Sequence Alignment , Trypsin Inhibitor, Kazal Pancreatic
19.
Dev Biol ; 275(1): 104-23, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15464576

ABSTRACT

In bilaterians, COUP-TF nuclear receptors participate in neurogenesis and/or CNS patterning. In hydra, the nervous system is formed of sensory mechanoreceptor cells (nematocytes) and neuronal cells, both lineages deriving from a common stem cell. The hydra COUP-TF gene, hyCOUP-TF, which encodes highly conserved DNA-binding and ligand-binding domains, belongs to the monophyletic COUP-TFs orphan receptor family (NR2F). In adult polyps, hyCOUP-TF is expressed in nematoblasts and a subset of neuronal cells. Comparative BrDU labeling analyses performed on cells expressing either hyCOUP-TF or the paired-like gene prdl-b showed that prdl-b expression corresponded to early stages of proliferation, while hyCOUP-TF was detected slightly later. HyCOUP-TF and prdl-b expressing cells disappeared in sf-1 mutants becoming "nerve-free". Moreover hyCOUP-TF and prdl-b expression was excluded from regions undergoing developmental processes. These data suggest that hyCOUP-TF and prdl-b belong to a genetic network that appeared together with neurogenesis during early metazoan evolution. The hyCOUP-TF protein specifically bound onto the evolutionarily conserved DR1 and DR5 response elements, and repressed transactivation induced by RAR:RXR nuclear receptors in a dose-dependent manner when expressed in mammalian cells. Hence, a cnidarian transcription factor can be active in vertebrate cells, implying that functional interactions between COUP-TF and other nuclear receptors were evolutionarily conserved.


Subject(s)
Cnidaria/metabolism , DNA-Binding Proteins/metabolism , Receptors, Steroid/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Biological Evolution , Biomarkers/metabolism , COUP Transcription Factors , Conserved Sequence , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , Homeodomain Proteins/metabolism , Hydra/metabolism , Molecular Sequence Data , Neurons/metabolism , Phylogeny , Protein Isoforms/genetics , Protein Isoforms/isolation & purification , Protein Isoforms/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/isolation & purification , Sequence Alignment , Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/isolation & purification
20.
Biosystems ; 76(1-3): 75-87, 2004.
Article in English | MEDLINE | ID: mdl-15351132

ABSTRACT

Cnidarians represent the first animal phylum with an organized nervous system and a complex active behavior. The hydra nervous system is formed of sensory-motoneurons, ganglia neurons and mechanoreceptor cells named nematocytes, which all differentiate from a common stem cell. The neurons are organized as a nerve net and a subset of neurons participate in a more complex structure, the nerve ring that was identified in most cnidarian species at the base of the tentacles. In order to better understand the genetic control of this neuronal network, we analysed the expression of evolutionarily conserved regulatory genes in the hydra nervous system. The Prd-class homeogene prdl-b and the nuclear orphan receptor hyCOUP-TF are expressed at strong levels in proliferating nematoblasts, a lineage where they were found repressed during patterning and morphogenesis, and at low levels in distinct subsets of neurons. Interestingly, Prd-class homeobox and COUP-TF genes are also expressed during neurogenesis in bilaterians, suggesting that mechanoreceptor and neuronal cells derive from a common ancestral cell. Moreover, the Prd-class homeobox gene prdl-a, the Antp-class homeobox gene msh, and the thrombospondin-related gene TSP1, which are expressed in distinct subset of neurons in the adult polyp, are also expressed during early budding and/or head regeneration. These data strengthen the fact that two distinct regulations, one for neurogenesis and another for patterning, already apply to these regulatory genes, a feature also identified in bilaterian related genes.


Subject(s)
Evolution, Molecular , Gene Expression Regulation, Developmental/physiology , Hydra/physiology , Nerve Regeneration/physiology , Nervous System/embryology , Nervous System/metabolism , Neurons/physiology , Transcription Factors/metabolism , Animals , Biological Evolution , Body Patterning/physiology , Hydra/cytology , Hydra/embryology , Morphogenesis , Nerve Net/cytology , Nerve Net/embryology , Nerve Net/physiology , Nervous System/cytology , Neurons/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...