Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(3): 113881, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38442019

ABSTRACT

An intriguing effect of short-term caloric restriction (CR) is the expansion of certain stem cell populations, including muscle stem cells (satellite cells), which facilitate an accelerated regenerative program after injury. Here, we utilized the MetRSL274G (MetRS) transgenic mouse to identify liver-secreted plasminogen as a candidate for regulating satellite cell expansion during short-term CR. Knockdown of circulating plasminogen prevents satellite cell expansion during short-term CR. Furthermore, loss of the plasminogen receptor KT (Plg-RKT) is also sufficient to prevent CR-related satellite cell expansion, consistent with direct signaling of plasminogen through the plasminogen receptor Plg-RKT/ERK kinase to promote proliferation of satellite cells. Importantly, we are able to replicate many of these findings in human participants from the CALERIE trial. Our results demonstrate that CR enhances liver protein secretion of plasminogen, which signals directly to the muscle satellite cell through Plg-RKT to promote proliferation and subsequent muscle resilience during CR.


Subject(s)
Plasminogen , Receptors, Cell Surface , Mice , Animals , Humans , Plasminogen/metabolism , Receptors, Cell Surface/metabolism , Caloric Restriction , Liver/metabolism , Mice, Transgenic , Serine Proteases , Cell Proliferation , Muscles/metabolism
2.
Mol Oncol ; 18(1): 113-135, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37971174

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly fatal metastatic disease associated with robust activation of the coagulation and fibrinolytic systems. However, the potential contribution of the primary fibrinolytic protease plasminogen to PDAC disease progression has remained largely undefined. Mice bearing C57Bl/6-derived KPC (KRasG12D , TRP53R172H ) tumors displayed evidence of plasmin activity in the form of high plasmin-antiplasmin complexes and high plasmin generation potential relative to mice without tumors. Notably, plasminogen-deficient mice (Plg- ) had significantly diminished KPC tumor growth in subcutaneous and orthotopic implantation models. Moreover, the metastatic potential of KPC cells was significantly diminished in Plg- mice, which was linked to reduced early adhesion and/or survival of KPC tumor cells. The reduction in primary orthotopic KPC tumor growth in Plg- mice was associated with increased apoptosis, reduced accumulation of pro-tumor immune cells, and increased local proinflammatory cytokine production. Elimination of fibrin(ogen), the primary proteolytic target of plasmin, did not alter KPC primary tumor growth and resulted in only a modest reduction in metastatic potential. In contrast, deficiencies in the plasminogen receptors Plg-RKT or S100A10 in tumor cells significantly reduced tumor growth. Plg-RKT reduction in tumor cells, but not reduced S100A10, suppressed metastatic potential in a manner that mimicked plasminogen deficiency. Finally, tumor growth was also reduced in NSG mice subcutaneously or orthotopically implanted with patient-derived PDAC tumor cells in which circulating plasminogen was pharmacologically reduced. Collectively, these studies suggest that plasminogen promotes PDAC tumor growth and metastatic potential, in part through engaging plasminogen receptors on tumor cells.


Subject(s)
Adenocarcinoma , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Humans , Mice , Carcinoma, Pancreatic Ductal/pathology , Fibrinolysin , Pancreatic Neoplasms/pathology , Plasminogen
3.
Adipocyte ; 12(1): 2252729, 2023 12.
Article in English | MEDLINE | ID: mdl-37642146

ABSTRACT

The plasminogen receptor, Plg-RKT, is a unique cell surface receptor that is broadly expressed in cells and tissues throughout the body. Plg-RKT localizes plasminogen on cell surfaces and promotes its activation to the broad-spectrum serine protease, plasmin. In this study, we show that overexpression of Plg-RKT protects mice from high fat diet (HFD)-induced adipose and metabolic dysfunction. During the first 10 weeks on the HFD, the body weights of mice that overexpressed Plg-RKT (Plg-RKT-OEX) were lower than those of control mice (CagRosaPlgRKT). After 10 weeks on the HFD, CagRosaPlgRKT and Plg-RKT-OEX mice had similar body weights. However, Plg-RKT-OEX mice showed a more metabolically favourable body composition phenotype. Plg-RKT-OEX mice also showed improved glucose tolerance and increased insulin sensitivity. We found that the improved metabolic functions of Plg-RKT-OEX mice were mechanistically associated with increased energy expenditure and activity, decreased proinflammatory adipose macrophages and decreased inflammation, elevated brown fat thermogenesis, and higher expression of adipose PPARγ and adiponectin. These findings suggest that Plg-RKT signalling promotes healthy adipose function via multiple mechanisms to defend against obesity-associated adverse metabolic phenotypes.


Subject(s)
Obesity , Serine Proteases , Animals , Mice , Mice, Obese , Obesity/etiology , Body Weight , Diet, High-Fat/adverse effects , Homeostasis , Plasminogen , Glucose
4.
JCI Insight ; 8(8)2023 04 24.
Article in English | MEDLINE | ID: mdl-36917195

ABSTRACT

Sepsis is a lethal syndrome characterized by systemic inflammation and abnormal coagulation. Despite therapeutic advances, sepsis mortality remains substantially high. Herein, we investigated the role of the plasminogen/plasmin (Plg/Pla) system during sepsis. Plasma levels of Plg were significantly lower in mice subjected to severe compared with nonsevere sepsis, whereas systemic levels of IL-6, a marker of sepsis severity, were higher in severe sepsis. Plg levels correlated negatively with IL-6 in both septic mice and patients, whereas plasminogen activator inhibitor-1 levels correlated positively with IL-6. Plg deficiency render mice susceptible to nonsevere sepsis induced by cecal ligation and puncture (CLP), resulting in greater numbers of neutrophils and M1 macrophages, liver fibrin(ogen) deposition, lower efferocytosis, and increased IL-6 and neutrophil extracellular trap (NET) release associated with organ damage. Conversely, inflammatory features, fibrin(ogen), and organ damage were substantially reduced, and efferocytosis was increased by exogenous Pla given during CLP- and LPS-induced endotoxemia. Plg or Pla protected mice from sepsis-induced lethality and enhanced the protective effect of antibiotics. Mechanistically, Plg/Pla-afforded protection was associated with regulation of NET release, requiring Pla-protease activity and lysine binding sites. Plg/Pla are important host-protective players during sepsis, controlling local and systemic inflammation and collateral organ damage.


Subject(s)
Extracellular Traps , Sepsis , Mice , Animals , Fibrinolysin , Plasminogen , Extracellular Traps/metabolism , Interleukin-6/metabolism , Inflammation/metabolism , Sepsis/metabolism , Fibrin/metabolism
5.
Biomolecules ; 12(4)2022 03 26.
Article in English | MEDLINE | ID: mdl-35454092

ABSTRACT

The plasminogen activation system regulates the activity of the serine protease, plasmin. The role of plasminogen receptors in cancer progression is being increasingly appreciated as key players in modulation of the tumor microenvironment. The interaction of plasminogen with cells to promote plasminogen activation requires the presence of proteins exposing C-terminal lysines on the cell surface. Plg-RKT is a structurally unique plasminogen receptor because it is an integral membrane protein that is synthesized with and binds plasminogen via a C-terminal lysine exposed on the cell surface. Here, we have investigated the expression of Plg-RKT in human breast tumors and human breast cancer cell lines. Breast cancer progression tissue microarrays were probed with anti-Plg-RKT mAB and we found that Plg-RKT is widely expressed in human breast tumors, that its expression is increased in tumors that have spread to draining lymph nodes and distant organs, and that Plg-RKT expression is most pronounced in hormone receptor (HR)-positive tumors. Plg-RKT was detected by Western blotting in human breast cancer cell lines. By flow cytometry, Plg-RKT cell surface expression was highest on the most aggressive tumor cell line. Future studies are warranted to address the functions of Plg-RKT in breast cancer.


Subject(s)
Breast Neoplasms , Receptors, Cell Surface , Breast Neoplasms/genetics , Cell Membrane/metabolism , Female , Humans , Plasminogen/metabolism , Receptors, Cell Surface/genetics , Serine Proteases , Tumor Microenvironment
6.
J Thromb Haemost ; 20(3): 742-754, 2022 03.
Article in English | MEDLINE | ID: mdl-34897983

ABSTRACT

BACKGROUND: Plg-RKT , a unique transmembrane plasminogen receptor, enhances the activation of plasminogen to plasmin, and localizes the proteolytic activity of plasmin on the cell surface. OBJECTIVES: We investigated the role of Plg-RKT in adipose function, metabolic homeostasis, and obesity. METHODS: We used adipose tissue (AT) sections from bariatric surgery patients and from high fat diet (HFD)-induced obese mice together with immunofluorescence and real-time polymerase chain reaction to study adipose expression of Plg-RKT . Mice genetically deficient in Plg-RKT and littermate controls fed a HFD or control low fat diet (LFD) were used to determine the role of Plg-RKT in insulin resistance, glucose tolerance, type 2 diabetes, and associated mechanisms including adipose inflammation, fibrosis, and ectopic lipid storage. The role of Plg-RKT in adipogenesis was determined using 3T3-L1 preadipocytes and primary cultures established from Plg-RKT -deficient and littermate control mice. RESULTS: Plg-RKT was highly expressed in both human and mouse AT, and its levels dramatically increased during adipogenesis. Plg-RKT -deficient mice, when fed a HFD, gained more weight, developed more hepatic steatosis, and were more insulin resistant/glucose intolerant than HFD-fed wild-type littermates. Mechanistically, these metabolic defects were linked with increased AT inflammation, AT macrophage and T-cell accumulation, adipose and hepatic fibrosis, and decreased insulin signaling in the AT and liver. Moreover, Plg-RKT regulated the expression of PPARγ and other adipogenic molecules, suggesting a novel role for Plg-RKT in the adipogenic program. CONCLUSIONS: Plg-RKT coordinately regulates multiple aspects of adipose function that are important to maintain efficient metabolic homeostasis.


Subject(s)
Adipose Tissue , Homeostasis , Receptors, Cell Surface , Adipose Tissue/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Dietary Fats/pharmacology , Fibrosis , Glucose Tolerance Test , Humans , Inflammation/metabolism , Insulin Resistance , Mice , Plasminogen/metabolism , Receptors, Cell Surface/metabolism
7.
Int J Mol Sci ; 22(4)2021 Feb 08.
Article in English | MEDLINE | ID: mdl-33567773

ABSTRACT

The ability of cells to promote plasminogen activation on their surfaces is now well recognized, and several distinct cell surface proteins have been demonstrated to function as plasminogen receptors. Here, we review studies demonstrating that plasminogen bound to cells, in addition to plasminogen directly bound to fibrin, plays a major role in regulating fibrin surveillance. We focus on the ability of specific plasminogen receptors on eukaryotic cells to promote fibrinolysis in the in vivo setting by reviewing data obtained predominantly in murine models. Roles for distinct plasminogen receptors in fibrin surveillance in intravascular fibrinolysis, immune cell recruitment in the inflammatory response, wound healing, and lactational development are discussed.


Subject(s)
Fibrin/metabolism , Fibrinolysis , Plasminogen/metabolism , Receptors, Cell Surface/metabolism , Animals , Humans
8.
Blood ; 137(2): 248-257, 2021 01 14.
Article in English | MEDLINE | ID: mdl-32842150

ABSTRACT

Plasminogen activation rates are enhanced by cell surface binding. We previously demonstrated that exogenous plasminogen binds to phosphatidylserine-exposing and spread platelets. Platelets contain plasminogen in their α-granules, but secretion of plasminogen from platelets has not been studied. Recently, a novel transmembrane lysine-dependent plasminogen receptor, Plg-RKT, has been described on macrophages. Here, we analyzed the pool of plasminogen in platelets and examined whether platelets express Plg-RKT. Plasminogen content of the supernatant of resting and collagen/thrombin-stimulated platelets was similar. Pretreatment with the lysine analog, ε-aminocaproic acid, significantly increased platelet-derived plasminogen (0.33 vs 0.08 nmol/108 platelets) in the stimulated supernatant, indicating a lysine-dependent mechanism of membrane retention. Lysine-dependent, platelet-derived plasminogen retention on thrombin and convulxin activated human platelets was confirmed by flow cytometry. Platelets initiated fibrinolytic activity in fluorescently labeled plasminogen-deficient clots and in turbidimetric clot lysis assays. A 17-kDa band, consistent with Plg-RKT, was detected in the platelet membrane fraction by western blotting. Confocal microscopy of stimulated platelets revealed Plg-RKT colocalized with platelet-derived plasminogen on the activated platelet membrane. Plasminogen exposure was significantly attenuated in thrombin- and convulxin-stimulated platelets from Plg-RKT-/- mice compared with Plg-RKT+/+ littermates. Membrane exposure of Plg-RKT was not dependent on plasminogen, as similar levels of the receptor were detected in plasminogen-/- platelets. These data highlight Plg-RKT as a novel plasminogen receptor in human and murine platelets. We show for the first time that platelet-derived plasminogen is retained on the activated platelet membrane and drives local fibrinolysis by enhancing cell surface-mediated plasminogen activation.


Subject(s)
Blood Platelets/metabolism , Plasminogen/metabolism , Platelet Activation/physiology , Receptors, Cell Surface/metabolism , Animals , Humans , Mice
9.
Cell Death Dis ; 11(12): 1054, 2020 12 12.
Article in English | MEDLINE | ID: mdl-33311441

ABSTRACT

Wound healing is a complex physiologic process that proceeds in overlapping, sequential steps. Plasminogen promotes fibrinolysis and potentiates the inflammatory response during wound healing. We have tested the hypothesis that the novel plasminogen receptor, Plg-RKT, regulates key steps in wound healing. Standardized burn wounds were induced in mice and time dependence of wound closure was quantified. Healing in Plg-RKT-/- mice was significantly delayed during the proliferation phase. Expression of inflammatory cytokines was dysregulated in Plg-RKT-/- wound tissue. Consistent with dysregulated cytokine expression, a significant delay in wound healing during the proliferation phase was observed in mice in which Plg-RKT was specifically deleted in myeloid cells. Following wound closure, the epidermal thickness was less in Plg-RKT-/- wound tissue. Paradoxically, deletion of Plg-RKT, specifically in keratinocytes, significantly accelerated the rate of healing during the proliferation phase. Mechanistically, only two genes were upregulated in Plg-RKT-/- compared with Plg-RKT+/+ wound tissue, filaggrin, and caspase 14. Both filaggrin and caspase 14 promote epidermal differentiation and decrease proliferation, consistent with more rapid wound closure and decreased epidermal thickness during the remodeling phase. Fibrin clearance was significantly impaired in Plg-RKT-/- wound tissue. Genetic reduction of fibrinogen levels to 50% completely abrogated the effect of Plg-RKT deletion on the healing of burn wounds. Remarkably, the effects of Plg-RKT deletion on cytokine expression were modulated by reducing fibrinogen levels. In summary, Plg-RKT is a new regulator participating in different phases of cutaneous burn wound healing, which coordinately plays a role in the interrelated responses of inflammation, keratinocyte migration, and fibrinolysis.


Subject(s)
Fibrinolysis , Inflammation/metabolism , Plasminogen/metabolism , Receptors, Cell Surface/metabolism , Skin/pathology , Wound Healing , Animals , Burns/genetics , Burns/pathology , Cell Proliferation/genetics , Epidermis/pathology , Fibrinogen/metabolism , Fibrinolysis/genetics , Gene Deletion , Gene Expression Regulation , Heterozygote , Inflammation/genetics , Keratinocytes/pathology , Mice, Inbred C57BL , Receptors, Cell Surface/genetics , Wound Healing/genetics
10.
J Thromb Haemost ; 18(10): 2468-2481, 2020 10.
Article in English | MEDLINE | ID: mdl-32662180

ABSTRACT

Plg-RKT is a structurally unique transmembrane plasminogen receptor with both N- and C-terminal domains exposed on the extracellular face of the cell. Its C-terminal lysine functions to tether plasminogen to cell surfaces. Overexpression of Plg-RKT increases cell surface plasminogen binding capacity while genetic deletion of Plg-RKT decreases plasminogen binding. Plasminogen binding to Plg-RKT results in promotion of plasminogen activation to the broad spectrum serine protease plasmin. This function is promoted by the physical association of Plg-RKT with the urokinase receptor (uPAR). Plg-RKT is broadly expressed in cells and tissues throughout the organism and its sequence is remarkably conserved phylogenetically. Plg-RKT also is required for lactation and, thus, is necessary for survival of the species. This review provides an overview of established and emerging functions of Plg-RKT and highlights major roles for Plg-RKT in both the initiation and resolution of inflammation. While the roles for Plg-RKT in the inflammatory response are predominantly plasmin(ogen)-dependent, its role in lactation requires both plasminogen-dependent and plasminogen-independent mechanisms. Furthermore, the functions of Plg-RKT are dependent on sex. In view of the broad tissue distribution of Plg-RKT , its role in a broad array of physiological and pathological processes should provide a fruitful area for future investigation.


Subject(s)
Fibrinolysin , Plasminogen , Cell Membrane , Female , Humans , Inflammation
11.
Article in English | MEDLINE | ID: mdl-32549050

ABSTRACT

t-PA has a widespread neuroendocrine distribution including prominent expression in chromaffin cells of the sympathoadrenal system. Chromaffin cell t-PA is sorted into catecholamine storage vesicles and co-released with catecholamines in response to sympathoadrenal activation, suggesting that catecholamine storage vesicles may serve as a reservoir for the rapid release of t-PA. Chromogranin A (CgA), a major core protein in secretory vesicles throughout the neuroendocrine system, may play a crucial role in targeting proteins into the regulated secretory pathway, by forming aggregated "granin" complexes to which other proteins destined for the regulated secretory vesicle bind and become separated from constitutively secreted proteins in the trans-Golgi network (TGN). Formation of such complexes is facilitated by conditions of the TGN (low pH, high Ca+2). We tested the hypothesis that t-PA interacts specifically with CgA and that this interaction is enhanced under conditions of the TGN. Immobilized t-PA was incubated with 125I-CgA. t-PA interacted specifically and saturably with CgA and the interaction was domain-specific, mediated by the EGF/finger and kringle 1 domains of t-PA and by a specific internal hydrophilic domain within CgA (KERTHQQKKHSSYEDELSEVL) as assessed by antibody and peptide competition studies. The interaction of t-PA with aggregated CgA complexes may play a role in the targeting of t-PA and its release from neurosecretory cells. These results may have broad implications for the regulation of local neurosecretory cell plasminogen activation under both normal physiological conditions and pathological conditions including cerebral ischemia.

12.
Front Immunol ; 10: 1458, 2019.
Article in English | MEDLINE | ID: mdl-31316511

ABSTRACT

Inflammation resolution is an active process that functions to restore tissue homeostasis. Clearance of apoptotic leukocytes by efferocytosis at inflammatory sites plays an important role in inflammation resolution and induces remarkable macrophage phenotypic and functional changes. Here, we investigated the effects of deletion of either plasminogen (Plg) or the Plg receptor, Plg-RKT, on the resolution of inflammation. In a murine model of pleurisy, the numbers of total mononuclear cells recruited to the pleural cavity were significantly decreased in both Plg-/- and Plg-RKT-/- mice, a response associated with decreased levels of the chemokine CCL2 in pleural exudates. Increased percentages of M1-like macrophages were determined in pleural lavages of Plg-/- and Plg-RKT-/- mice without significant changes in M2-like macrophage percentages. In vitro, Plg and plasmin (Pla) increased CD206/Arginase-1 expression and the levels of IL-10/TGF-ß (M2 markers) while decreasing IFN/LPS-induced M1 markers in murine bone-marrow-derived macrophages (BMDMs) and human macrophages. Furthermore, IL4-induced M2-like polarization was defective in BMDMs from both Plg-/- and Plg-RKT-/- mice. Mechanistically, Plg and Pla induced transient STAT3 phosphorylation, which was decreased in Plg-/- and Plg-RKT-/- BMDMs after IL-4 or IL-10 stimulation. The extents of expression of CD206 and Annexin A1 (important for clearance of apoptotic cells) were reduced in Plg-/- and Plg-RKT-/- macrophage populations, which exhibited decreased phagocytosis of apoptotic neutrophils (efferocytosis) in vivo and in vitro. Taken together, these results suggest that Plg and its receptor, Plg-RKT, regulate macrophage polarization and efferocytosis, as key contributors to the resolution of inflammation.


Subject(s)
Macrophages/immunology , Plasminogen/immunology , Pleurisy/immunology , Receptors, Cell Surface/immunology , Animals , Cell Movement , Humans , Male , Mice, Transgenic , Neutrophils/immunology , Phagocytosis , Phenotype , Plasminogen/genetics , Receptors, Cell Surface/genetics
13.
Blood ; 134(6): 561-567, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31221672

ABSTRACT

Membrane-bound plasmin is used by immune cells to degrade extracellular matrices, which facilitates migration. The plasminogen receptor Plg-RKT is expressed by immune cells, including monocytes and macrophages. Among monocytes and macrophages, distinct subsets can be distinguished based on cell surface markers and pathophysiological function. We investigated expression of Plg-RKT by monocyte and macrophage subsets and whether potential differential expression might have functional consequences for cell migration. Proinflammatory CD14++CD16+ human monocytes and Ly6Chigh mouse monocytes expressed the highest levels of Plg-RKT and bound significantly more plasminogen compared with the other respective subsets. Proinflammatory human macrophages, generated by polarization with lipopolysaccharide and interferon-γ, showed significantly higher expression of Plg-RKT compared with alternatively activated macrophages, polarized with interleukin-4 and interleukin-13. Directional migration of proinflammatory monocytes was plasmin dependent and was abolished by anti-Plg-RKT monoclonal antibody, ε-amino-caproic acid, aprotinin, and the aminoterminal fragment of urokinase-type plasminogen activator. In an in vivo peritonitis model, significantly less Ly6Chigh monocyte recruitment was observed in Plg-RKT -/- compared with Plg-RKT +/+ mice. Immunohistochemical analysis of human carotid plaques and adipose tissue showed that proinflammatory macrophages also exhibited high levels of Plg-RKT in vivo. Our data demonstrate higher expression of Plg-RKT on proinflammatory monocyte and macrophage subsets that impacts their migratory capacity.


Subject(s)
Gene Expression Regulation , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Receptors, Cell Surface/genetics , Animals , Biomarkers , Cell Movement/immunology , Extracellular Matrix/metabolism , Humans , Immunophenotyping , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Mice
14.
15.
Cell Discov ; 3: 17001, 2017.
Article in English | MEDLINE | ID: mdl-28417010

ABSTRACT

Plasminogen activation is involved in many processes within the central nervous system, including synaptic plasticity, neuroinflammation and neurodegeneration. However, the mechanisms that regulate plasminogen activation in the brain still remain unknown. Here we demonstrate that astrocytes participate in this regulation by two mechanisms. First, the astrocyte plasma membrane serves as a surface for plasminogen activation by tissue-type plasminogen activator. This activation triggers downstream plasmin-dependent processes with important impacts in brain health and disease, such as fibrinolysis and brain-derived neurotrophic factor conversion. Second, astrocytes take up plasminogen and plasmin in a regulated manner through a novel mechanism involving endocytosis mediated by cell-surface actin and triggered by extracellular plasmin activity at the surface of astrocytes. Following endocytosis, plasminogen and plasmin are targeted to lysosomes for degradation. Thus, cell-surface actin acts as a sensor of plasmin activity to induce a negative feedback through plasmin endocytosis. This study provides evidence that astrocytes control the balance between plasmin formation and plasmin elimination in the brain parenchyma.

16.
Blood ; 127(9): 1079-80, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26941388

ABSTRACT

In this issue of Blood, Motley et al have identified a novel and unexpected mechanism for clearance of extravascular fibrin that is accomplished by a specific proinflammatory macrophage population and is dependent upon active plasmin, yet independent of known fibrinogen receptors.


Subject(s)
Endocytosis , Fibrin/metabolism , Macrophages/metabolism , Receptors, CCR2/metabolism , Animals
17.
Blood ; 124(5): 665-6, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25082858

ABSTRACT

In this issue of Blood, Das et al assign a very novel and unanticipated function to plasminogen by showing that it is an enhancer of the phagocytic function of macrophages.


Subject(s)
Kupffer Cells/metabolism , Macrophages, Peritoneal/metabolism , Phagocytosis/physiology , Plasminogen/metabolism , Animals
18.
Int Rev Cell Mol Biol ; 309: 259-302, 2014.
Article in English | MEDLINE | ID: mdl-24529725

ABSTRACT

Plasminogen (PLG) is the zymogen of plasmin, the major enzyme that degrades fibrin clots. In addition to its binding and activation on fibrin clots, PLG also specifically interacts with cell surfaces where it is more efficiently activated by PLG activators, compared with the reaction in solution. This results in association of the broad-spectrum proteolytic activity of plasmin with cell surfaces that functions to promote cell migration. Here, we review emerging data establishing a role for PLG, plasminogen receptors and the newly discovered plasminogen receptor, Plg-RKT, in macrophage recruitment in the inflammatory response, and we address mechanisms by which the interplay between PLG and its receptors regulates inflammation.


Subject(s)
Macrophages/metabolism , Plasminogen/metabolism , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Humans , Macrophages/pathology , Molecular Sequence Data , Peritonitis/pathology , Plasminogen/chemistry , Proteomics , Receptors, Cell Surface/chemistry
19.
Semin Thromb Hemost ; 39(4): 329-37, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23532575

ABSTRACT

The interaction of plasminogen with cell surfaces results in promotion of plasmin formation and retention on the cell surface. This results in arming cell surfaces with the broad-spectrum proteolytic activity of plasmin. Over the past quarter century, key functional consequences of the association of plasmin with the cell surface have been elucidated. Physiologic and pathophysiologic processes with plasmin-dependent cell migration as a central feature include inflammation, wound healing, oncogenesis, metastasis, myogenesis, and muscle regeneration. Cell surface plasmin also participates in neurite outgrowth and prohormone processing. Furthermore, plasmin-induced cell signaling also affects the functions of inflammatory cells, via production of cytokines, reactive oxygen species, and other mediators. Finally, plasminogen receptors regulate fibrinolysis. In this review, we highlight emerging data that shed light on longstanding controversies and raise new issues in the field. We focus on (1) the impact of the recent X-ray crystal structures of plasminogen and the development of antibodies that recognize cell-induced conformational changes in plasminogen on our understanding of the interaction of plasminogen with cells; (2) the relationship between apoptosis and plasminogen binding to cells; (3) the current status of our understanding of the molecular identity of plasminogen receptors and the discovery of a structurally unique novel plasminogen receptor, Plg-RKT; (4) the determinants of the interplay between distinct plasminogen receptors and cellular functions; and (5) new insights into the role of colocalization of plasminogen and plasminogen activator receptors on the cell surface.


Subject(s)
Plasminogen/metabolism , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Fibrinolysin/metabolism , Fibrinolysis/physiology , Humans , Molecular Sequence Data , Plasminogen Activators/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...