Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Nat Commun ; 13(1): 3117, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35660741

ABSTRACT

Intestinal stem cells (ISCs) at the crypt base contribute to intestinal homeostasis through a balance between self-renewal and differentiation. However, the molecular mechanisms regulating this homeostatic balance remain elusive. Here we show that the matricellular protein CCN1/CYR61 coordinately regulates ISC proliferation and differentiation through distinct pathways emanating from CCN1 interaction with integrins αvß3/αvß5. Mice that delete Ccn1 in Lgr5 + ISCs or express mutant CCN1 unable to bind integrins αvß3/αvß5 exhibited exuberant ISC expansion and enhanced differentiation into secretory cells at the expense of absorptive enterocytes in the small intestine, leading to nutrient malabsorption. Analysis of crypt organoids revealed that through integrins αvß3/αvß5, CCN1 induces NF-κB-dependent Jag1 expression to regulate Notch activation for differentiation and promotes Src-mediated YAP activation and Dkk1 expression to control Wnt signaling for proliferation. Moreover, CCN1 and YAP amplify the activities of each other in a regulatory loop. These findings establish CCN1 as a niche factor in the intestinal crypts, providing insights into how matrix signaling exerts overarching control of ISC homeostasis.


Subject(s)
Integrins , Intestines , Animals , Cell Differentiation , Cell Proliferation , Cysteine-Rich Protein 61/metabolism , Intestinal Mucosa , Mice , Wnt Signaling Pathway
2.
Nat Commun ; 11(1): 1242, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32144270

ABSTRACT

Expression of the matricellular protein CCN1 (CYR61) is associated with inflammation and is required for successful wound repair. Here, we show that CCN1 binds bacterial pathogen-associated molecular patterns including peptidoglycans of Gram-positive bacteria and lipopolysaccharides of Gram-negative bacteria. CCN1 opsonizes methicillin-resistant Staphylococcus aureus (MRSA) and Pseudomonas aeruginosa and accelerates their removal by phagocytosis and increased production of bactericidal reactive oxygen species in macrophages through the engagement of integrin αvß3. Mice with myeloid-specific Ccn1 deletion and knock-in mice expressing CCN1 unable to bind αvß3 are more susceptible to infection by S. aureus or P. aeruginosa, resulting in increased mortality and organ colonization. Furthermore, CCN1 binds directly to TLR2 and TLR4 to activate MyD88-dependent signaling, cytokine expression and neutrophil mobilization. CCN1 is therefore a pattern recognition receptor that opsonizes bacteria for clearance and functions as a damage-associated molecular pattern to activate inflammatory responses, activities that contribute to wound healing and tissue repair.


Subject(s)
Cysteine-Rich Protein 61/metabolism , Opsonin Proteins/metabolism , Pseudomonas Infections/immunology , Staphylococcal Infections/immunology , Toll-Like Receptors/metabolism , Animals , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/immunology , Disease Models, Animal , Disease Susceptibility , Female , Gene Knock-In Techniques , Gene Knockdown Techniques , Humans , Integrin alphaVbeta3/immunology , Integrin alphaVbeta3/metabolism , Male , Methicillin-Resistant Staphylococcus aureus , Mice , Mice, Inbred C57BL , Mice, Transgenic , Opsonin Proteins/genetics , Pathogen-Associated Molecular Pattern Molecules/metabolism , Phagocytosis/immunology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Sf9 Cells , Signal Transduction/immunology , Staphylococcal Infections/microbiology , Toll-Like Receptors/immunology
3.
J Clin Invest ; 128(1): 97-107, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29293097

ABSTRACT

Fibrosis is the excessive accumulation of extracellular matrix that often occurs as a wound healing response to repeated or chronic tissue injury, and may lead to the disruption of organ architecture and loss of function. Although fibrosis was previously thought to be irreversible, recent evidence indicates that certain circumstances permit the resolution of fibrosis when the underlying causes of injury are eradicated. The mechanism of fibrosis resolution encompasses degradation of the fibrotic extracellular matrix as well as elimination of fibrogenic myofibroblasts through their adaptation of various cell fates, including apoptosis, senescence, dedifferentiation, and reprogramming. In this Review, we discuss the present knowledge and gaps in our understanding of how matrix degradation is regulated and how myofibroblast cell fates can be manipulated, areas that may identify potential therapeutic approaches for fibrosis.


Subject(s)
Apoptosis , Cell Dedifferentiation , Cellular Reprogramming , Cellular Senescence , Extracellular Matrix/metabolism , Myofibroblasts/metabolism , Animals , Extracellular Matrix/pathology , Fibrosis , Humans , Myofibroblasts/pathology
4.
Nat Commun ; 6: 7386, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26077348

ABSTRACT

Neutrophil infiltration constitutes the first step in wound healing, although their timely clearance by macrophage engulfment, or efferocytosis, is critical for efficient tissue repair. However, the specific mechanism for neutrophil clearance in wound healing remains undefined. Here we uncover a key role for CCN1 in neutrophil efferocytosis by acting as a bridging molecule that binds phosphatidylserine, the 'eat-me' signal on apoptotic cells and integrins αvß3/αvß5 in macrophages to trigger efferocytosis. Both knockin mice expressing a mutant CCN1 that is unable to bind αvß3/αvß5 and mice with Ccn1 knockdown are defective in neutrophil efferocytosis, resulting in exuberant neutrophil accumulation and delayed healing. Treatment of wounds with CCN1 accelerates neutrophil clearance in both Ccn1 knockin mice and diabetic Lepr(db/db) mice, which suffer from neutrophil persistence and impaired healing. These findings establish CCN1 as a critical opsonin in skin injury and suggest a therapeutic potential for CCN1 in certain types of non-healing wounds.


Subject(s)
Cysteine-Rich Protein 61/genetics , Cytophagocytosis/genetics , Macrophages/immunology , Neutrophils/immunology , Skin/injuries , Wound Healing/genetics , Animals , Cell Migration Assays , Cysteine-Rich Protein 61/immunology , Cysteine-Rich Protein 61/pharmacology , Cytophagocytosis/drug effects , Cytophagocytosis/immunology , Diabetes Mellitus/genetics , Diabetes Mellitus/immunology , Epidermal Growth Factor/pharmacology , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Keratinocytes/drug effects , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Real-Time Polymerase Chain Reaction , Receptors, Leptin/genetics , Receptors, Vitronectin , Reverse Transcriptase Polymerase Chain Reaction , Skin/drug effects , Skin/immunology , Wound Healing/drug effects , Wound Healing/immunology
5.
Nat Rev Drug Discov ; 10(12): 945-63, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22129992

ABSTRACT

Members of the CCN family of matricellular proteins are crucial for embryonic development and have important roles in inflammation, wound healing and injury repair in adulthood. Deregulation of CCN protein expression or activities contributes to the pathobiology of various diseases - many of which may arise when inflammation or tissue injury becomes chronic - including fibrosis, atherosclerosis, arthritis and cancer, as well as diabetic nephropathy and retinopathy. Emerging studies indicate that targeting CCN protein expression or signalling pathways holds promise in the development of diagnostics and therapeutics for such diseases. This Review summarizes the biology of CCN proteins, their roles in various pathologies and their potential as therapeutic targets.


Subject(s)
CCN Intercellular Signaling Proteins/metabolism , Drug Delivery Systems/trends , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Animals , CCN Intercellular Signaling Proteins/chemistry , Disease Models, Animal , Fibrosis , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology
6.
Aging (Albany NY) ; 2(9): 627-31, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20930261

ABSTRACT

Mammalian wound healing involves the rapid synthesis and deposition of extracellular matrix (ECM) to maintain tissue integrity during repair. This process must be tightly controlled, as its deregulation may result in fibrosis, scarring, and loss of tissue function. Recent studies have uncovered an efficient and parsimonious mechanism for rendering fibrogenesis self-limiting in wound healing: in such diverse organs as the liver and skin, the myofibroblasts that initially proliferate and produce ECM are themselves eventually driven into senescence, blocking their further proliferation and converting them into matrix-degrading cells. Myofibroblast senescence in skin wounds is triggered by a dynamically expressed matricellular protein, CCN1/CYR61, which acts through integrin-mediated induction of oxidative stress. We propose that the onset of myofibroblast senescence is a programmed wound healing response that functions as a self-limiting mechanism for fibrogenesis, and this process may be regulated by the ECM microenvironment through the expression of CCN1/CYR61.


Subject(s)
Cellular Senescence/physiology , Skin/pathology , Wound Healing/physiology , Cell Proliferation , Cysteine-Rich Protein 61/physiology , Extracellular Matrix/physiology , Fibrosis/physiopathology , Humans , Myofibroblasts/cytology , Oxidative Stress/physiology , Skin/physiopathology
7.
Nat Cell Biol ; 12(7): 676-85, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20526329

ABSTRACT

Cellular senescence is a recognized mechanism of tumour suppression; however, its contribution to other pathologies is not well understood. We show that the matricellular protein CCN1 (also known as CYR61; cysteine-rich protein 61), which is dynamically expressed at sites of wound repair, can induce fibroblast senescence by binding to integrin alpha(6)beta(1) and the heparan sulphate proteoglycans (receptors involved in cell adhesion). CCN1 induces DNA damage response pathways and activates p53 and the RAC1-NOX1 complex, which generates reactive oxygen species (ROS). This results in the ROS-dependent activation of the p16(INK4a)/pRb pathway, leading to senescence and concomitant expression of antifibrotic genes. Senescent fibroblasts accumulate in granulation tissues of healing cutaneous wounds and express antifibrotic genes in wild-type mice. These processes are lost in knockin mice that express a senescence-defective Ccn1 mutant, resulting in exacerbated fibrosis. Topical application of CCN1 protein to wounds reverses these defects. Thus, fibroblast senescence is a CCN1-dependent wound healing response in cutaneous injury that functions to curb fibrosis during tissue repair.


Subject(s)
Cellular Senescence/drug effects , Cysteine-Rich Protein 61/pharmacology , Wound Healing/drug effects , Animals , Blotting, Western , Cell Line , Cell Proliferation/drug effects , Collagen/metabolism , Fibroblasts , Humans , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , RNA Interference , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
8.
Proc Natl Acad Sci U S A ; 106(36): 15326-31, 2009 Sep 08.
Article in English | MEDLINE | ID: mdl-19706414

ABSTRACT

Osteopontin (OPN) is highly expressed in cancer patients and plays important roles in many stages of tumor progression, such as anti-apoptosis, proliferation, and metastasis. From functional screening of human cDNA library, we isolated OPN as a caspase-8 substrate that regulates cell death during hypoxia/reoxygenation (Hyp/RO). In vitro cleavage assays demonstrate that OPN is cleaved at Asp-135 and Asp-157 by caspase-8. Cellular cleavage of OPN is observed in apoptotic cells exposed to Hyp/RO among various apoptotic stimuli and its cleavage is blocked by zVAD or IETD caspase inhibitor. Further, over-expression of OPN, the form with secretion signal, inhibits Hyp/RO-induced cell death. Caspase cleavage-defective OPN mutant (OPN D135A/D157A) is more efficient to suppress Hyp/RO-induced cell death than wild-type OPN. OPN D135A/D157A sustains AKT activity to increase cell viability through inhibition of caspase-9 during Hyp/RO. In addition, OPN is highly induced in some tumor cells during Hyp/RO, such as HeLa and Huh-7 cells, which is associated with their resistance to Hyp/RO by sustaining AKT activity. Notably, OPN C-terminal cleavage fragment produced by caspase-8 is detected in the nucleus. Plasmid-encoded expression of OPN C-terminal cleavage fragment increases p53 protein level and induces apoptosis of wild-type mouse embryonic fibroblast cells, but not p53(-/-) mouse embryonic fibroblast cells. These observations suggest that the protective function of OPN during Hyp/RO is inactivated via the proteolytic cleavage by caspase-8 and its cleavage product subsequently induces cell death via p53, postulating caspase-8 as a negative regulator of tumorigenic activity of OPN.


Subject(s)
Apoptosis/physiology , Caspase 8/metabolism , Cell Hypoxia/physiology , Osteopontin/metabolism , Tumor Suppressor Protein p53/metabolism , Blotting, Western , Densitometry , HeLa Cells , Humans , Osteopontin/genetics
9.
J Biol Chem ; 280(21): 20722-9, 2005 May 27.
Article in English | MEDLINE | ID: mdl-15778222

ABSTRACT

Autophagic cell death is characterized by the accumulation of vacuoles in physiological and pathological conditions. However, its molecular event is unknown. Here, we show that Atg5, which is known to function in autophagy, contributes to autophagic cell death by interacting with Fas-associated protein with death domain (FADD). Down-regulation of Atg5 expression in HeLa cells suppresses cell death and vacuole formation induced by IFN-gamma. Inversely, ectopic expression of Atg5 using adenoviral delivery induces autophagic cell death. Deletion mapping analysis indicates that procell death activity resides in the middle and C-terminal region of Atg5. Cells harboring the accumulated vacuoles triggered by IFN-gamma or Atg5 expression become dead, and vacuole formation precedes cell death. 3-Methyladenine or expression of Atg5(K130R) mutant blocks both cell death and vacuole formation triggered by IFN-gamma, whereas benzyloxycarbonyl-VAD-fluoromethyl ketone (Z-VAD-fmk) inhibits only cell death but not vacuole formation. Atg5 interacts with FADD via death domain in vitro and in vivo, and the Atg5-mediated cell death, but not vacuole formation, is blocked in FADD-deficient cells. These results suggest that Atg5 plays a crucial role in IFN-gamma-induced autophagic cell death by interacting with FADD.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Adenine/analogs & derivatives , Autophagy/physiology , Cell Death/physiology , Microtubule-Associated Proteins/physiology , Vacuoles/physiology , Adaptor Proteins, Signal Transducing/deficiency , Adenine/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Autophagy-Related Protein 5 , Binding Sites , Cell Death/drug effects , Cell Line , Fas-Associated Death Domain Protein , Gene Deletion , Gene Expression , HeLa Cells , Humans , Interferon-gamma/pharmacology , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Mutagenesis , Oligonucleotides, Antisense/pharmacology , Saccharomyces cerevisiae , Transfection , Two-Hybrid System Techniques , Vacuoles/drug effects
10.
Oncogene ; 24(4): 688-96, 2005 Jan 20.
Article in English | MEDLINE | ID: mdl-15592525

ABSTRACT

Caspase-8 is the most receptor-proximal, upstream caspase in the caspase cascade and plays a key role in cell death triggered by various death receptors. Here, we addressed the role of endogenous caspase-8 in tumor necrosis factor (TNF)-alpha-induced activation of NF-kappaB. Direct targeting of caspase-8 with siRNA and antisense (AS) approaches abolished TNF-alpha-induced activation of NF-kappaB in NIH3T3, HeLa, and HEK293 cells as determined with luciferase reporter gene and cell fractionation assays. Reconstitution of caspase-8-deficient C33A cells with processing-defective (P/D) mutant of caspase-8 sensitized the cells to TNF-alpha for NF-kappaB activation. In contrast to wild-type caspase-8, death effector domain mutant replacing Asp73 with Ala (caspase-8 (D73A)) failed to activate NF-kappaB and to bind FLICE-associated huge protein (FLASH) in vitro and in vivo. Instead, caspase-8 (D73A) mutant bound to caspase-8 and blocked NF-kappaB activation triggered by TNF-alpha and caspase-8. In addition, expression of an NF-kappaB-activating domain-deletion mutant of FLASH or transfection of FLASH AS oligonucleotides abolished TNF-alpha and caspase-8, but not phorbol 12-myristate 13-acetate, -induced activation of NF-kappaB. Further, immunoprecipitation assays showed that caspase-8 formed triple complex with TRAF2 and FLASH. Taken together, these results suggest that endogenous caspase-8 mediates TNF-alpha-induced activation of NF-kappaB via FLASH.


Subject(s)
Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Caspases/metabolism , Mutation/genetics , NF-kappa B/metabolism , Signal Transduction , Animals , Apoptosis Regulatory Proteins , Aspartic Acid/genetics , Aspartic Acid/metabolism , Caspase 8 , Caspase Inhibitors , Caspases/deficiency , Caspases/genetics , Cell Line , Enzyme Inhibitors/pharmacology , Humans , Mice , Protein Binding , Signal Transduction/drug effects , TNF Receptor-Associated Factor 2/metabolism , Tumor Necrosis Factor-alpha/pharmacology
11.
Mol Cell Biol ; 24(22): 9763-70, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15509781

ABSTRACT

Apoptosis repressor with CARD (ARC) possesses the ability not only to block activation of caspase 8 but to modulate caspase-independent mitochondrial events associated with cell death. However, it is not known how ARC modulates both caspase-dependent and caspase-independent cell death. Here, we report that ARC is a Ca(2+)-dependent regulator of caspase 8 and cell death. We found that in Ca(2+) overlay and Stains-all assays, ARC protein bound to Ca(2+) through the C-terminal proline/glutamate-rich (P/E-rich) domain. ARC expression reduced not only cytosolic Ca(2+) transients but also cytotoxic effects of thapsigargin, A23187, and ionomycin, for which the Ca(2+)-binding domain of ARC was indispensable. Conversely, direct interference of endogenous ARC synthesis by targeting ARC enhanced such Ca(2+)-mediated cell death. In addition, binding and immunoprecipitation analyses revealed that the protein-protein interaction between ARC and caspase 8 was decreased by the increase of Ca(2+) concentration in vitro and by the treatment of HEK293 cells with thapsigargin in vivo. Caspase 8 activation was also required for the thapsigargin-induced cell death and suppressed by the ectopic expression of ARC. These results suggest that calcium binding mediates regulation of caspase 8 and cell death by ARC.


Subject(s)
Apoptosis/physiology , Calcium/metabolism , Caspases/metabolism , Muscle Proteins/metabolism , Animals , Apoptosis Regulatory Proteins , COS Cells , Caspase 8 , Cell Line , Enzyme Activation/drug effects , HeLa Cells , Humans , In Vitro Techniques , Jurkat Cells , Muscle Proteins/chemistry , Muscle Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thapsigargin/pharmacology
12.
FEBS Lett ; 578(3): 239-44, 2004 Dec 17.
Article in English | MEDLINE | ID: mdl-15637807

ABSTRACT

We have identified a novel CARD-containing protein from EST database. BinCARD (Bcl10-interacting protein with CARD). BinCARD was ubiquitously expressed. Co-immunoprecipitation, In vitro binding, mammalian two-hybrid, and immunostaining assays revealed that BinCARD interacted with Bcl10 through CARD. BinCARD potently suppressed NF-kappa B activation induced by Bcl10 and decreased the amounts of phosphorylated Bcl10. Mutations at the residue Leu17 or Leu65, which is highly conserved in CARD, abolished the inhibitory effects of BinCARD on both Bcl10-induced activation of NF-kappa B and phosphorylation of Bcl10. Further, expression of BinCARD inhibited Bcl10 phosphorylation induced by T cell activation signal. These results suggest that BinCARD interacts with Bcl10 to inhibit Bcl10-mediated activation of NF-kappa B and to suppress Bcl10 phosphorylation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , CARD Signaling Adaptor Proteins/metabolism , Carrier Proteins/metabolism , NF-kappa B/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Motifs , Amino Acid Sequence , B-Cell CLL-Lymphoma 10 Protein , Blotting, Western , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Conserved Sequence , DNA Mutational Analysis , Down-Regulation , Glutathione Transferase/metabolism , HeLa Cells , Humans , Immunohistochemistry , Jurkat Cells , Leucine/chemistry , Leucine/genetics , Luciferases/metabolism , Lymphocyte Activation , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Precipitin Tests , Protein Binding , Protein Structure, Secondary , RNA, Messenger/metabolism , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , T-Lymphocytes/metabolism , Transcription, Genetic , Two-Hybrid System Techniques
13.
J Biol Chem ; 278(26): 24003-10, 2003 Jun 27.
Article in English | MEDLINE | ID: mdl-12702723

ABSTRACT

FAF1 has been introduced as a Fas-binding protein. However, the function of FAF1 in apoptotic execution is not established. Based on the fact that FAF1 is a Fas-binding protein, we asked if FAF1 interacted with other members of the Fas-death-inducing signaling complex (Fas-DISC) such as Fas-associated death domain protein (FADD) and caspase-8. FAF1 could interact with caspase-8 and FADD in vivo as well as in vitro. The death effector domains (DEDs) of caspase-8 and FADD interacted with the amino acid 181-381 region of FAF1, previously known to have apoptotic potential. Considering that FAF1 directly binds to Fas and caspase-8, FAF1 shows similar protein-interacting characteristics to that of FADD. In the coimmunoprecipitation with an anti-Fas antibody (APO-1) in Jurkat cells, endogenous FAF1 was associated with the precipitates in which caspase-8 was present. By confocal microscopic analysis, both Fas and FAF1 were detected in the cytoplasmic membrane before Fas activation, and in the cytoplasm after Fas activation. FADD and caspase-8 colocalized with Fas in Jurkat cells validating the presence of FAF1 in the authentic Fas-DISC. Overexpression of FAF1 in Jurkat cells caused significant apoptotic death. In addition, the FAF1 deletion mutant lacking the N terminus where Fas, FADD, and caspase-8 interact protected Jurkat cells from Fas-induced apoptosis demonstrating dominant-negative phenotype. Cell death by overexpression of FAF1 was suppressed significantly in both FADD- and caspase-8-deficient Jurkat cells when compared with that in their parental Jurkat cells. Collectively, our data show that FAF1 is a member of Fas-DISC acting upstream of caspase-8.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Amino Acid Sequence , Apoptosis , Apoptosis Regulatory Proteins , Caspase 8 , Caspase 9 , Caspases/metabolism , Death Domain Receptor Signaling Adaptor Proteins , Fas-Associated Death Domain Protein , Humans , Jurkat Cells , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Alignment , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL