Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Clin Appl Thromb Hemost ; 28: 10760296221077096, 2022.
Article in English | MEDLINE | ID: mdl-35166576

ABSTRACT

Thrombomodulin alfa (TM-α, recombinant human soluble thrombomodulin) and antithrombin (AT) concentrate are anticoagulant agents for the treatment of disseminated intravascular coagulation (DIC). A post hoc analysis using data from 1198 patients with infection-induced DIC from the post-marketing surveillance of TM-α was conducted. To identify subgroups that benefit from combination therapy, the patients were a priori stratified into four groups by a platelet (Plt) count of 50 × 103/µL and plasma AT level of 50% (groups 1, 2, 3, and 4, with high Plt/high AT, high Plt/low AT, low Plt/high AT, and low Plt/low AT, respectively). Kaplan-Meier survival analysis showed significantly worse survival in groups 2 and 4 had than in group 1 (p = 0.0480, p < 0.0001, respectively), and multivariate analysis showed that concomitant AT concentrate was independently correlated with reduced 28-day mortality only in group 4 (hazard ratio 0.6193; 95% confidence interval, 0.3912-0.9805). The adverse drug reactions (ADRs) and bleeding ADRs were not different among the groups. Patients with both severe thrombocytopenia and AT deficiency are candidates for combined anticoagulant therapy with TM-α and AT concentrate.


Subject(s)
Anticoagulants/therapeutic use , Antithrombins/therapeutic use , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Sepsis/complications , Thrombomodulin/therapeutic use , Aged , Aged, 80 and over , Anticoagulants/administration & dosage , Antithrombins/administration & dosage , Antithrombins/adverse effects , Disseminated Intravascular Coagulation/mortality , Drug Therapy, Combination , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Platelet Count , Thrombomodulin/administration & dosage
2.
Biochem Biophys Res Commun ; 587: 1-8, 2022 01 08.
Article in English | MEDLINE | ID: mdl-34856423

ABSTRACT

BACKGROUND: Accidental hypothermia (AH) sometimes leads to coagulation disorder, especially in severe AH. We previously demonstrated that intrasplenic platelet activation caused aberrant hemostasis and thrombus formation after rewarming in a murine AH model. However, no study has focused on the appropriate management of platelets causing coagulation activation after rewarming of AH. We investigated whether or not recombinant soluble thrombomodulin (rTM) can suppress thrombosis formation after rewarming using a rat AH model. METHODS: Wistar rats were exposed to an ambient temperature of -20 °C under general anesthesia until their rectal temperature decreased to 26 °C. The Hypo group rats (n = 5) were immediately euthanized, while the Hypo/Re group (n = 5) and rTM group rats (n = 5), which were administered rTM (1 mg/kg) via the tail vein, were rewarmed until the rectal temperature returned to 34 °C and then euthanized 6 h later. Tissue and blood samples were collected from all rats for histopathological and coagulation analyses at euthanasia. RESULTS: There was no significant change in the D-dimer level in the Hypo group rats, while the D-dimer level was significantly elevated at 6 h after rewarming in the Hypo/Re group rats (P = 0.015), and histopathology detected both fibrin and platelets in the renal glomerulus. However, the rTM group rats did not show any elevation of the D-dimer levels at 6 h after rewarming, and no fibrin was noted on histopathology. CONCLUSIONS: rTM may be useful as an appropriate anticoagulant in cases of aberrant hemostasis after rewarming of AH.


Subject(s)
Anticoagulants/pharmacology , Blood Platelets/drug effects , Fibrin Fibrinogen Degradation Products/metabolism , Hypothermia/complications , Thrombomodulin/administration & dosage , Thrombosis/prevention & control , Animals , Biomarkers/metabolism , Blood Platelets/metabolism , Blood Platelets/pathology , Disease Models, Animal , Fibrin/chemistry , Fibrin/metabolism , Hypothermia/blood , Hypothermia/physiopathology , Kidney Glomerulus/blood supply , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Platelet Activation/drug effects , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Rewarming/adverse effects , Solubility , Spleen/blood supply , Spleen/drug effects , Spleen/metabolism , Spleen/pathology , Thrombosis/blood , Thrombosis/etiology , Thrombosis/physiopathology
3.
Cells ; 10(9)2021 08 29.
Article in English | MEDLINE | ID: mdl-34571886

ABSTRACT

Diabetes mellitus is a global threat to human health. The ultimate cause of diabetes mellitus is insufficient insulin production and secretion associated with reduced pancreatic ß-cell mass. Apoptosis is an important and well-recognized mechanism of the progressive loss of functional ß-cells. However, there are currently no available antiapoptotic drugs for diabetes mellitus. This study evaluated whether recombinant human thrombomodulin can inhibit ß-cell apoptosis and improve glucose intolerance in a diabetes mouse model. A streptozotocin-induced diabetes mouse model was prepared and treated with thrombomodulin or saline three times per week for eight weeks. The glucose tolerance and apoptosis of ß-cells were evaluated. Diabetic mice treated with recombinant human thrombomodulin showed significantly improved glucose tolerance, increased insulin secretion, decreased pancreatic islet areas of apoptotic ß-cells, and enhanced proportion of regulatory T cells and tolerogenic dendritic cells in the spleen compared to counterpart diseased mice treated with saline. Non-diabetic mice showed no changes. This study shows that recombinant human thrombomodulin, a drug currently used to treat patients with coagulopathy in Japan, ameliorates glucose intolerance by protecting pancreatic islet ß-cells from apoptosis and modulating the immune response in diabetic mice. This observation points to recombinant human thrombomodulin as a promising antiapoptotic drug for diabetes mellitus.


Subject(s)
Apoptosis/drug effects , Blood Glucose/drug effects , Diabetes Mellitus, Experimental/prevention & control , Hypoglycemic Agents/administration & dosage , Islets of Langerhans/drug effects , Thrombomodulin/administration & dosage , Animals , Biomarkers/blood , Blood Glucose/metabolism , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Injections, Intraperitoneal , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/administration & dosage , Spleen/drug effects , Spleen/immunology , Spleen/metabolism , Streptozocin , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
4.
J Pharmacol Sci ; 146(1): 49-57, 2021 May.
Article in English | MEDLINE | ID: mdl-33858655

ABSTRACT

We performed clinical retrospective study in female cancer patients and fundamental experiments in mice, in order to clarify risk factors for paclitaxel-induced peripheral neuropathy (PIPN). In the clinical study, 131 of 189 female outpatients with cancer undergoing paclitaxel-based chemotherapy met inclusion criteria. Breast cancer survivors (n = 40) showed significantly higher overall PIPN (grades 1-4) incidence than non-breast cancer survivors (n = 91). Multivariate sub-analyses of breast cancer survivors showed that 57 years of age or older and endocrine therapy before paclitaxel treatment were significantly associated with severe PIPN (grades 2-4). The age limit was also significantly correlated with overall development of severe PIPN. In the preclinical study, female mice subjected to ovariectomy received repeated administration of paclitaxel, and mechanical nociceptive threshold was assessed by von Frey test. Ovariectomy aggravated PIPN in the mice, an effect prevented by repeated treatment with 17ß-estradiol. Repeated administration of thrombomodulin alfa (TMα), known to prevent chemotherapy-induced peripheral neuropathy in rats and mice, also prevented the development of PIPN in the ovariectomized mice. Collectively, breast cancer survivors, particularly with postmenopausal estrogen decline and/or undergoing endocrine therapy, are considered a PIPN-prone subpopulation, and may require non-hormonal pharmacological intervention for PIPN in which TMα may serve as a major candidate.


Subject(s)
Antineoplastic Agents, Phytogenic/adverse effects , Estrogens/deficiency , Estrogens/physiology , Paclitaxel/adverse effects , Peripheral Nervous System Diseases/chemically induced , Age Factors , Aged , Aged, 80 and over , Animals , Breast Neoplasms , Cancer Survivors , Female , Humans , Mice , Mice, Inbred Strains , Middle Aged , Ovariectomy/adverse effects , Peripheral Nervous System Diseases/prevention & control , Postmenopause , Rats , Retrospective Studies , Risk Factors , Thrombomodulin/administration & dosage
5.
Medicine (Baltimore) ; 100(9): e25044, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33655983

ABSTRACT

RATIONALE: Chronic disseminated intravascular coagulation (DIC) associated with thoracic aortic aneurysm is characterized by enhanced fibrinolysis and is thought to be stable in the compensated/asymptomatic stage, with few bleeding symptoms. However, DIC can lead to decompensated/hemorrhagic stage disseminated intravascular coagulation, resulting in severe bleeding diathesis, and there is currently no established strategy for treatment of DIC in aortic aneurysms. PATIENT CONCERNS: A 77-year-old woman underwent angiography and cardiac catheterization, before descending aortic replacement surgery. She developed DIC in postprocedure week 2 with extensive, uncontrollable massive subcutaneous hemorrhage. DIAGNOSES: Her acute-phase DIC score was 7 points, and the risk of mortality within 30 days after surgery according to the JapanSCORE was estimated to be 33.6%. INTERVENTIONS: Therapy was a combination of recombinant human soluble thrombomodulin (rhTM) and an aortic stent-graft treatment. OUTCOMES: Short-term improvements were seen in both DIC and bleeding diathesis. The thoracic aortic aneurysm with severe DIC was eventually corrected by administration of rhTM. LESSONS: We report the use of rhTM as an effective, novel anticoagulant drug with anti-inflammatory activity for treating DIC with suppressed fibrinolysis, which is typically associated with sepsis. In patients with a high hemorrhagic diathesis, in whom preoperative control of DIC cannot be achieved with conventional anticoagulation and radical surgical repair cannot be performed, a combination of rhTM and endovascular therapy may be a powerful new treatment option.


Subject(s)
Aortic Aneurysm, Thoracic/etiology , Disseminated Intravascular Coagulation/drug therapy , Preoperative Care/methods , Thrombomodulin/administration & dosage , Vascular Surgical Procedures , Aged , Angiography , Aortic Aneurysm, Thoracic/diagnosis , Aortic Aneurysm, Thoracic/surgery , Disseminated Intravascular Coagulation/complications , Dose-Response Relationship, Drug , Female , Humans , Injections, Intravenous , Recombinant Proteins/administration & dosage
6.
Endocrinology ; 162(4)2021 04 01.
Article in English | MEDLINE | ID: mdl-33406229

ABSTRACT

Preeclampsia (PE) is a common gestational complication that involves systemic endothelial dysfunction and inflammatory responses primarily due to placental damage. Recombinant thrombomodulin (rTM), a novel anticoagulant clinically used for disseminated intravascular coagulation, is reported to have a unique anti-inflammatory endothelial repair function by inhibiting proinflammatory mediator high-mobility group box 1 (HMGB1). Despite the severe patient outcomes, there are currently no effective therapeutic options to treat PE. Here, we verified the efficacy of rTM as a novel therapeutic agent for PE using a murine model and human trophoblast cells. We revealed the therapeutic potential of rTM in an angiotensin II(Ang II)-induced PE mouse model. Injection of rTM significantly attenuated clinical features of PE, such as hypertension, proteinuria, fetal growth restriction, and impaired placental vasculature. Elevation of maternal soluble fms-like tyrosine kinase-1 (sFlt-1), a well-accepted causal factor of PE that induces systemic endothelial dysfunction, was suppressed in response to rTM treatment. Supporting these findings, our in vitro experiments revealed that rTM reduces Ang II-triggered overproduction of sFlt-1 in human trophoblast cells. Moreover, interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), well-known key inflammatory mediators in PE pathogenesis, were diminished by rTM. SiRNA knockdown experiments further determined that these processes were directly mediated by HMGB1. Our studies demonstrate that rTM exerts its clinical effect as HMBG1 inhibitor and ameliorates placental dysfunction, which is central to PE pathogenesis. Our findings suggest that rTM could be a promising therapeutic that significantly improve the outcomes of PE patients.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , HMGB1 Protein/antagonists & inhibitors , Pre-Eclampsia/drug therapy , Thrombomodulin/administration & dosage , Animals , Disease Models, Animal , Female , HMGB1 Protein/genetics , HMGB1 Protein/immunology , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Mice, Inbred C57BL , Placenta/drug effects , Placenta/immunology , Pre-Eclampsia/genetics , Pre-Eclampsia/immunology , Pregnancy , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Thrombomodulin/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
7.
Cancer Lett ; 497: 1-13, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33065249

ABSTRACT

Surgery is the only curative treatment option for pancreatic cancer, but patients often develop postoperative recurrence. Surgical invasiveness might be involved in the mechanism of recurrence. The associations among inflammation caused by surgery, neutrophils, and cancer metastasis were investigated. At first, neutrophil extracellular traps (NETs) were examined in clinical specimens, and NETs were observed around metastatic tumors. To explore how NETs were induced, neutrophils were cultured with pancreatic cancer or in cancer-conditioned medium. Neutrophils formed NETs when they were cultured with pancreatic cancer or even its conditioned medium. The effects of NETs on cancer cells were further investigated in vitro and in vivo. NETs induced the epithelial to mesenchymal transition in cancer cells and thereby promoted their migration and invasion. HMGB1 derived from NETs appeared to potentiate the malignancy of cancer cells. In a mouse model of liver metastasis with inflammation, NETs participated in the metastatic process by enhancing extravasation. Interestingly, thrombomodulin degraded HMGB1 and consequently inhibited the induction of NETs, thereby preventing pancreatic cancer metastasis to the liver. In conclusion, NETs interact reciprocally with pancreatic cancer cells, which play a pivotal role in inflammation-associated metastasis. Targeting NETs with thrombomodulin can be a novel strategy to improve the surgical outcome of pancreatic cancer patients.


Subject(s)
Disease Models, Animal , Extracellular Traps/metabolism , Liver Neoplasms/prevention & control , Neutrophils/metabolism , Pancreatic Neoplasms/prevention & control , Reperfusion Injury/prevention & control , Thrombomodulin/metabolism , Animals , Apoptosis , Carcinogens/toxicity , Cell Proliferation , Epithelial-Mesenchymal Transition , Humans , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Mice , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Tetradecanoylphorbol Acetate/toxicity , Thrombomodulin/administration & dosage , Thrombomodulin/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Int J Hematol ; 113(1): 15-23, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33175341

ABSTRACT

Aortic aneurysms and vascular malformations are sometimes associated with disseminated intravascular coagulation (DIC). A typical blood coagulation test shows decrease in platelet count and fibrinogen, and increases in fibrin/fibrinogen degradation products (FDP) and D-dimer. The coagulation activation marker thrombin-antithrombin complex (TAT) and the fibrinolysis activation marker plasmin-α2 plasmin inhibitor (PIC) are significantly increased. α2 plasmin inhibitor (α2PI) is significantly reduced. Since no prolongation of prothrombin time (PT) is noticeable and activated partial thromboplastin time (APTT) is shortened in some cases, DIC cannot be diagnosed or ruled out by PT and APTT alone. The cornerstone of treatment for DIC is to treat the underlying disease. However, surgery is not possible in some cases. Follow-up may be appropriate in patients with abnormal results from coagulation tests and no bleeding. However, pharmacotherapy is often required in cases with bleeding. Unfractionated heparin, low molecular weight heparin, protease inhibitors, recombinant thrombomodulin, direct oral anticoagulants, and factor XIII preparations are effective. If PIC is significantly increased and α2PI is significantly decreased, or if the bleeding is severe, tranexamic acid is used as an antifibrinolytic therapy with anticoagulant therapy. In such cases, attention should be paid not only to TAT but also changes in PIC.


Subject(s)
Anticoagulants/administration & dosage , Antifibrinolytic Agents/administration & dosage , Antithrombin III/analysis , Aortic Aneurysm/complications , Blood Vessels/abnormalities , Disseminated Intravascular Coagulation/diagnosis , Disseminated Intravascular Coagulation/etiology , Fibrinolysin/analysis , Peptide Hydrolases/analysis , Tranexamic Acid/administration & dosage , alpha-2-Antiplasmin/analysis , Administration, Oral , Aged , Aged, 80 and over , Biomarkers/blood , Disseminated Intravascular Coagulation/drug therapy , Female , Heparin, Low-Molecular-Weight/administration & dosage , Humans , Male , Partial Thromboplastin Time , Protease Inhibitors/administration & dosage , Prothrombin Time , Thrombomodulin/administration & dosage
9.
Cancer Chemother Pharmacol ; 86(5): 607-618, 2020 11.
Article in English | MEDLINE | ID: mdl-32965539

ABSTRACT

PURPOSE: The purpose of this clinical study was to be the first to explore whether ART-123, a recombinant human soluble thrombomodulin, prevents oxaliplatin-induced peripheral neuropathy (OIPN). METHODS: This randomized, phase IIa trial enrolled stage II/III colon cancer patients who received adjuvant mFOLFOX6 chemotherapy. Participants were randomly allocated to 3 arms in a double-blind manner: placebo (placebo: days 1-3); 1-day ART (ART-123: day 1, placebo: days 2-3); and 3-day ART (ART-123: days 1-3). ART-123 (380 U/kg/day) or placebo was infused intravenously before each 2-week cycle of mFOLFOX6. OIPN was assessed with the Functional Assessment of Cancer Therapy/Gynecological Oncology Group-Neurotoxicity-12 (FACT/GOG-Ntx-12) score by participants and the NCI Common Terminology Criteria for Adverse Events (NCI-CTCAE) by investigators. RESULTS: Seventy-nine participants (placebo n = 28, 1-day ART n = 27, 3-day ART n = 24) received study drugs. The least-squares mean FACT/GOG-Ntx-12 scores at cycle 12 from the mixed effect model for repeated measures were 28.9 with placebo, 36.3 with 1-day ART (vs. placebo: 7.3 [95% CI 1.9 to12.8, p = 0.009]), and 32.3 with 3-day ART (vs. placebo: 3.4 [95% CI -.1 to 9.0, p = 0.222]). The cumulative incidence of NCI-CTCAE grade ≥ 2 sensory neuropathy at cycle 12 was 64.3% with placebo, 40.7% with 1-day ART (vs. placebo: -23.5 [95% CI -48.4 to 4.0], p = 0.108), and 45.8% with 3-day ART (vs. placebo: -18.5 [95% CI -44.2 to 9.4], p = 0.264). Common adverse events were consistent with those reported with mFOLFOX6; no severe bleeding adverse events occurred. CONCLUSION: ART-123 showed a potential preventive effect against OIPN with good tolerability. A larger study with 1-day ART is warranted. NCT02792842, registration date: June 8, 2016.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Colonic Neoplasms/drug therapy , Oxaliplatin/adverse effects , Peripheral Nervous System Diseases/prevention & control , Thrombomodulin/administration & dosage , Adult , Aged , Drug Administration Schedule , Female , Fluorouracil/adverse effects , Humans , Incidence , Infusions, Intravenous , Leucovorin/adverse effects , Male , Middle Aged , Organoplatinum Compounds/adverse effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/epidemiology , Placebos , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Self Report/statistics & numerical data , Severity of Illness Index , Treatment Outcome
10.
J Surg Res ; 256: 536-542, 2020 12.
Article in English | MEDLINE | ID: mdl-32799002

ABSTRACT

Antithrombin deficiency (ATD) was described in 1965 by Olav Egeberg as the first known inherited form of thrombophilia. Today, it is understood that ATDs can be congenital or acquired, leading to qualitative, quantitative, or mixed abnormalities in antithrombin (AT). All ATDs ultimately hinder AT's ability to serve as an endogenous anticoagulant and antiinflammatory agent. As a result, ATD patients possess higher risk for thromboembolism and can develop recurrent venous and arterial thromboses. Because heparin relies on AT to augment its physiologic function, patients with ATD often exhibit profound heparin resistance. Although rare as a genetic disorder, acquired forms of ATD are seen with surprising frequency in critically ill patients. This review discusses ATD in the context of surgical critical care with specific relevance to trauma, thermal burns, cardiothoracic surgery, and sepsis.


Subject(s)
Anticoagulants/administration & dosage , Antithrombin III Deficiency/complications , Postoperative Complications/prevention & control , Surgical Procedures, Operative/adverse effects , Thrombosis/prevention & control , Administration, Oral , Antithrombin III/genetics , Antithrombin III Deficiency/diagnosis , Antithrombin III Deficiency/drug therapy , Antithrombin III Deficiency/genetics , Antithrombins/administration & dosage , Antithrombins/metabolism , Critical Care , Critical Illness , Heparin/metabolism , Humans , Mutation , Postoperative Complications/etiology , Recombinant Proteins/administration & dosage , Sepsis/blood , Sepsis/complications , Sepsis/surgery , Thrombomodulin/administration & dosage , Thrombosis/etiology , Wounds and Injuries/blood , Wounds and Injuries/complications , Wounds and Injuries/surgery
11.
J Neuroinflammation ; 17(1): 240, 2020 Aug 14.
Article in English | MEDLINE | ID: mdl-32799887

ABSTRACT

BACKGROUND: Excessive inflammation within damaged tissue usually leads to delayed or insufficient regeneration, and nerves in the peripheral nervous system (PNS) generally do not recover fully following damage. Consequently, there is growing interest in whether modulation of the inflammatory response could help to promote nerve regeneration in the PNS. However, to date, there are no practical therapeutic strategies for manipulating inflammation after nerve injury. Thrombomodulin (TM) is a transmembrane glycoprotein containing five domains. The lectin-like domain of TM has the ability to suppress the inflammatory response. However, whether TM can modulate inflammation in the PNS during nerve regeneration has yet to be elucidated. METHODS: We investigated the role of TM in switching proinflammatory type 1 macrophages (M1) to anti-inflammatory type 2 macrophages (M2) in a human monocytic cell line (THP-1) and evaluated the therapeutic application of TM in transected sciatic nerve injury in rats. RESULTS: The administration of TM during M1 induction significantly reduced the expression levels of inflammatory cytokines, including TNF-a (p < 0.05), IL-6 (p < 0.05), and CD86 (p < 0.05), in THP-1 cells. Simultaneously, the expression levels of M2 markers, including IL-10 (p < 0.05) and CD206 (p < 0.05), were significantly increased in TM-treated THP-1 cells. Inhibition of IL-4R-c-Myc-pSTAT6-PPARγ signaling abolished the expression levels of IL-10 (p < 0.05) and CD206 (p < 0.05). The conditioned medium (CM) collected from M1 cells triggered an inflammatory response in primary Schwann cells, while CM collected from M1 cells treated with TM resulted in a dose-dependent reduction in inflammation. TM treatment led to better nerve regeneration when tested 6 weeks after injury and preserved effector muscle function. In addition, TM treatment reduced macrophage infiltration at the site of injury and led to potent M1 to M2 transition, thus indicating the anti-inflammatory capacity of TM. CONCLUSIONS: Collectively, our findings demonstrate the anti-inflammatory role of TM during nerve regeneration. Therefore, TM represents a potential drug for the promotion and modulation of functional recovery in peripheral nerves that acts by regulating the M1/M2 ratio.


Subject(s)
Macrophages/drug effects , Nerve Regeneration/drug effects , Peripheral Nerve Injuries/drug therapy , Peripheral Nerves/drug effects , Thrombomodulin/administration & dosage , Animals , Cell Line , Cell Polarity/drug effects , Cytokines/metabolism , Humans , Macrophages/metabolism , Male , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/physiopathology , Peripheral Nerves/metabolism , Peripheral Nerves/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
12.
Mol Cell Biochem ; 475(1-2): 195-203, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32767229

ABSTRACT

Regulatory T cells (Tregs) and transforming growth factor ß (TGF-ß) are believed to play key roles in both postoperative pro-inflammatory and anti-inflammatory responses of malignancies. Recombinant human thrombomodulin (rTM) is implied to inhibit the interaction between TGF-ß and Tregs. The aim of this study is to evaluate the antitumor effects of rTM against gastrointestinal tumors under systemic inflammation. Mice were subjected to cecal ligation and puncture and percutaneous allogeneic tumor implantation. rTM were introduced by percutaneous injection into the abdominal cavity. The effects of rTM were evaluated by weight of implanted tumor, proportion of Tregs in peripheral blood lymphocytes (PBL) and tumor infiltrating lymphocytes (TIL) and temporal evaluation of serum cytokines. The effect of rTM was also evaluated on the in vitro differentiation of naïve T cells into induced Tregs induced by TGF-ß and interleukin (IL) -2. rTM significantly inhibited the proliferation of the implanted tumor cells in an inflammation-dependent manner. rTM also reduced the fractions of regulatory T cells and induced regulatory T cells among both PBL and TIL. Temporal evaluation of serum cytokine levels in the model mice showed that rTM significantly suppressed the increases in the serum levels of IL-2 and TGF-ß. An in vitro differentiation assay revealed that rTM inhibited the differentiation of naïve T cells into Tregs triggered by IL-2- and TGF-ß. rTM has suppressive effects on inflammation-induced gastrointestinal tumor growth by suggestively affecting differentiation of Tregs.


Subject(s)
Gastrointestinal Neoplasms/drug therapy , Inflammation/complications , Peritonitis/complications , T-Lymphocytes, Regulatory/immunology , Thrombomodulin/administration & dosage , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Disease Models, Animal , Female , Gastrointestinal Neoplasms/etiology , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Mice , Mice, Inbred BALB C , Recombinant Proteins/administration & dosage
13.
Cells ; 9(8)2020 07 30.
Article in English | MEDLINE | ID: mdl-32751580

ABSTRACT

Endothelial cellular stiffening has been observed not only in inflamed cultured endothelial cells but also in the endothelium of atherosclerotic regions, which is an underlying cause of monocyte adhesion and accumulation. Although recombinant soluble thrombomodulin (rsTM) has been reported to suppress the inflammatory response of endothelial cells, its role in regulating endothelial cellular stiffness remains unclear. The purpose of this study was to investigate the impact of anticoagulant rsTM on lipopolysaccharide (LPS)-induced endothelial cellular stiffening. We show that LPS increases endothelial cellular stiffness by using atomic force microscopy and that rsTM reduces LPS-induced cellular stiffening not only through the attenuation of actin fiber and focal adhesion formation but also via the improvement of gap junction functionality. Moreover, post-administration of rsTM, after LPS stimulation, attenuated LPS-induced cellular stiffening. We also found that endothelial cells regulate leukocyte adhesion in a substrate- and cellular stiffness-dependent manner. Our result show that LPS-induced cellular stiffening enhances monocytic THP-1 cell line adhesion, whereas rsTM suppresses THP-1 cell adhesion to inflamed endothelial cells by reducing cellular stiffness. Endothelial cells increase cellular stiffness in reaction to inflammation, thereby promoting monocyte adhesion. Treatment of rsTM reduced LPS-induced cellular stiffening and suppressed monocyte adhesion in a cellular stiffness-dependent manner.


Subject(s)
Actins/ultrastructure , Cell Adhesion/drug effects , Focal Adhesions/drug effects , Gap Junctions/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Lipopolysaccharides/pharmacology , Monocytes/metabolism , Thrombomodulin/administration & dosage , Thrombomodulin/chemistry , Anticoagulants/administration & dosage , Anticoagulants/chemistry , Atherosclerosis/metabolism , Focal Adhesions/ultrastructure , Gap Junctions/ultrastructure , Humans , Inflammation/drug therapy , Microscopy, Atomic Force , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Signal Transduction/drug effects , Solubility , THP-1 Cells
15.
In Vivo ; 34(3): 1037-1045, 2020.
Article in English | MEDLINE | ID: mdl-32354890

ABSTRACT

AIM: The present study aimed to examine the effects of prophylactic administration of recombinant human soluble thrombomodulin (rTM) for the prevention of sinusoidal obstruction syndrome (SOS). MATERIALS AND METHODS: Crl:CD1 mice were allocated to the rTM, placebo, and control groups. The rTM group received an intraperitoneal administration of rTM, with intraperitoneal administration of monocrotaline (MCT) 1 h later. The placebo group received PBS instead of rTM, and the control group received PBS instead of rTM and MCT. Mice were sacrificed 48 h after MCT administration, and blood and liver tissues were evaluated. Immunostaining was performed using anti-CD42b and anti-SE-1 antibodies, and AZAN staining. Levels of plasminogen activator inhibitor (PAI-1) and endothelial nitric oxide synthase (eNOS) in whole liver tissues were estimated using RT-PCR. RESULTS: Hematoxylin-eosin staining showed that SOS-related findings were markedly attenuated in the rTM group compared to the placebo group. CD42b immunostaining showed the presence of extravasated platelet activation (EPA) in the Disse space in the placebo group, but this was less noticeable in the rTM group. PAI-1 levels were significantly lower in the rTM group than in the placebo group in RT-PCR. However, eNOS levels were significantly higher in the rTM group than in the placebo group. CONCLUSION: Administration of rTM may prevent SOS by protecting sinusoidal endothelial cells.


Subject(s)
Hepatic Veno-Occlusive Disease/prevention & control , Recombinant Proteins/administration & dosage , Thrombomodulin/administration & dosage , Animals , Biomarkers , Biopsy , Blood Cell Count , Disease Models, Animal , Hepatic Veno-Occlusive Disease/diagnosis , Hepatic Veno-Occlusive Disease/etiology , Humans , Immunohistochemistry , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Pre-Exposure Prophylaxis , Recombinant Proteins/therapeutic use , Thrombomodulin/therapeutic use , Treatment Outcome
16.
Sci Rep ; 10(1): 4874, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32184456

ABSTRACT

Septic patients can develop disseminated intravascular coagulation (DIC), which is characterized by systemic blood coagulation and an increased risk of life-threatening haemorrhage. Although antithrombin (AT) and thrombomodulin (TM) combination anticoagulant therapy is frequently used to treat septic patients with DIC in Japan, its effectiveness in improving patient outcomes remains unclear. In this large-scale multicentre retrospective study of adult septic patients with DIC treated at Japanese hospitals between February 2010 and March 2016, we compared in-hospital mortality between AT monotherapy and AT + TM combination therapy. We performed logistic regression analysis with in-hospital mortality as the dependent variable and anticoagulant therapy as the main independent variable of interest. Covariates included patient demographics, disease severity, and body surface area. The AT group and AT + TM group comprised 1,017 patients from 352 hospitals and 1,205 patients from 349 hospitals, respectively. AT + TM combination therapy was not significantly associated with lower mortality when compared with AT monotherapy (odds ratio: 0.97, 95% confidence interval: 0.78-1.21; P = 0.81). AT + TM combination therapy was also not superior to AT monotherapy in reducing mechanical ventilation or hospitalization durations. Despite its widespread use for treating sepsis with DIC, AT + TM combination therapy is not more effective in improving prognoses than the simpler AT monotherapy.


Subject(s)
Antithrombins/administration & dosage , Disseminated Intravascular Coagulation/drug therapy , Respiration, Artificial/methods , Sepsis/therapy , Thrombomodulin/administration & dosage , Aged , Aged, 80 and over , Antithrombins/therapeutic use , Disseminated Intravascular Coagulation/etiology , Disseminated Intravascular Coagulation/mortality , Drug Therapy, Combination , Female , Hospital Mortality , Humans , Japan , Male , Middle Aged , Retrospective Studies , Sepsis/complications , Sepsis/mortality , Thrombomodulin/therapeutic use , Treatment Outcome
17.
PLoS One ; 15(1): e0228093, 2020.
Article in English | MEDLINE | ID: mdl-31971961

ABSTRACT

Multiple organ dysfunction induced by sepsis often involves kidney injury. Extracellular histones released in response to damage-associated molecular patterns are known to facilitate sepsis-induced organ dysfunction. Recombinant human soluble thrombomodulin (rhTM) and its lectin-like domain (D1) exert anti-inflammatory effects and neutralize damage-associated molecular patterns. However, the effects of rhTM and D1 on extracellular histone H3 levels and kidney injury remain poorly understood. Our purpose was to investigate the association between extracellular histone H3 levels and kidney injury, and to clarify the effects of rhTM and D1 on extracellular histone H3 levels, kidney injury, and survival in sepsis-induced rats. Rats in whom sepsis was induced via cecal ligation and puncture were used in this study. Histone H3 levels, histopathology of the kidneys, and the survival rate of rats at 24 h after cecal ligation and puncture were investigated. Histone H3 levels increased over time following cecal ligation and puncture. Histopathological analyses indicated that the distribution of degeneration foci among tubular epithelial cells of the kidney and levels of histone H3 increased simultaneously. Administration of rhTM and D1 significantly reduced histone H3 levels compared with that in the vehicle-treated group and improved kidney injury. The survival rates of rats in rhTM- and D1-treated groups were significantly higher than that in the vehicle-treated group. The results of this study indicated that rhTM and its D1 similarly reduce elevated histone H3 levels, thereby reducing acute kidney injury. Our findings also proposed that rhTM and D1 show potential as new treatment strategies for sepsis combined with acute kidney injury.


Subject(s)
Extracellular Space/metabolism , Histones/metabolism , Kidney/pathology , Recombinant Proteins/therapeutic use , Sepsis/drug therapy , Thrombomodulin/therapeutic use , Animals , Cecum/pathology , Creatinine/blood , Histones/blood , Humans , Kidney/drug effects , Ligation , Male , Protein Domains , Punctures , Rats, Wistar , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Sepsis/blood , Survival Analysis , Thrombomodulin/administration & dosage , Thrombomodulin/chemistry
18.
Gen Thorac Cardiovasc Surg ; 68(8): 785-792, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31955320

ABSTRACT

OBJECTIVES: Systemic inflammation evoked by cardiopulmonary bypass (CPB) leads to acute lung injury (ALI) and respiratory failure. Although recombinant human soluble thrombomodulin (rTM) consists of three domains (D1-3), is reported to attenuate systemic inflammation through the N-terminal lectin-like domain (D1), anticoagulant domain (D2) may exacerbate coagulopathy after CPB. We investigated the potential of selective D1 against CPB-mediated ALI free from anticoagulant effects using a rat CPB model. METHODS: Rats were divided into three groups: control (CPB alone, n = 5), D1 (CPB + D1, n = 4), and D123 (CPB + D123, n = 6). D1 or D123 was administrated to the rats of each group before CPB establishment. Blood samples are collected before, during and after CPB. Blood coagulability was assessed by a coagulation analyzer. Lung samples are collected at 1 h after the termination of CPB for histological analyses. RESULTS: D123 group exhibited significantly prolonged glass beads-activated clotting time with heparinase after CPB compared to that in control group, whereas no significant prolongation in control and D1 group (control vs. D1 vs. D123: 65.4 ± 9.2 vs. 65.3 ± 10.9 vs. 83.5 ± 4.6 s, p = 0.036 [control vs. D123], 0.99 [control vs. D1]) indicating the absence of anticoagulant activities of D1. Histological studies revealed less congestion, edema, inflammation, and hemorrhage in both D1 and D123 groups compared to those in control group indicating protective effects of both D1 and D123 against ALI mediated by CPB. CONCLUSIONS: N-terminal lectin-like domain of rTM may reduce the risk of ALI without anticoagulant effects.


Subject(s)
Acute Lung Injury/prevention & control , Anticoagulants/therapeutic use , Cardiopulmonary Bypass/adverse effects , Thrombomodulin/therapeutic use , Acute Lung Injury/etiology , Animals , Anticoagulants/administration & dosage , Disease Models, Animal , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Thrombomodulin/administration & dosage
19.
Eur J Pharm Sci ; 139: 105066, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31513922

ABSTRACT

Thrombomodulin (TM) is an endothelial cell membrane protein that plays essential roles in controlling vascular haemostatic balance. The 4, 5, 6 EGF-like domain of TM (TM456) has cofactor activity for thrombin binding and subsequently protein C activation. Therefore, recombinant TM456 is a promising anticoagulant candidate but has a very short half-life. Ligation of poly (ethylene glycol) to a bioactive protein (PEGylation) is a practical choice to improve stability, extend circulating life, and reduce immunogenicity of the protein. Site-specific PEGylation is preferred as it could avoid the loss of protein activity resulting from nonspecific modification. We report herein two site-specific PEGylation strategies, enzymatic ligation and copper-free click chemistry (CFCC), for rTM456 modification. Recombinant TM456 with a C-terminal LPETG tag (rTM456-LPETG) was expressed in Escherichia coli for its end-point modification with NH2-diglycine-PEG5000-OMe via Sortase A-mediated ligation (SML). Similarly, an azide functionality was easily introduced at the C-terminus of rTM456-LPETG via SML with NH2-diglycine-PEG3-azide, which facilitates a site-specific PEGylation of rTM456via CFCC. Both PEGylated rTM456 conjugates retained protein C activation activity as that of rTM456. Also, they were more stable than rTM456 in Trypsin digestion assay. Further, both PEGylated rTM456 conjugates showed a concentration-dependent prolongation of thrombin clotting time (TCT) compared to non-modified protein, which confirms the effectiveness of these two site-specific PEGylation schemes.


Subject(s)
Anticoagulants/administration & dosage , Anticoagulants/chemistry , Thrombomodulin/administration & dosage , Thrombomodulin/chemistry , Azides/administration & dosage , Azides/chemistry , Blood Coagulation/drug effects , Click Chemistry , Drug Stability , Humans , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Thrombin/metabolism , Thrombomodulin/genetics
20.
Surg Today ; 49(11): 971-976, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31190184

ABSTRACT

PURPOSE: Necrotizing enterocolitis (NEC) remains the leading cause of death in preterm infants. Recombinant human soluble thrombomodulin (rTM) has been reported to have anti-inflammatory effects as well as antithrombogenic effects. The aim of this study was to evaluate the effect of rTM in a rat NEC model. METHODS: NEC was induced by enteral feeding with hyperosmolar formula, gavage administration of lipopolysaccharide and asphyxia stress. Controls were fed by their mother ad libitum. In the treatment group, rTM was administered subcutaneously twice (once each on the first and second day). All animals surviving beyond 96 h or that developed signs of distress were euthanized. The ileum was harvested for a histological evaluation and the measurement of the mRNA and protein expression. RESULTS: The rate of NEC-like intestinal injury in the treatment group (9/25, 36%) was significantly lower than in the NEC group (25/34, 73.5%). Tissue levels of TNF-α, IL-6 and HMGB1 were significantly elevated in the NEC group, whereas those in the treatment group were decreased to similar values as in the control group. CONCLUSIONS: Our experimental study showed that rTM is able to reduce the severity and incidence of NEC. It may be an alternative option for the treatment of NEC.


Subject(s)
Enterocolitis, Necrotizing/prevention & control , Thrombomodulin/administration & dosage , Animals , Animals, Newborn , Biomarkers/metabolism , Disease Models, Animal , Enterocolitis, Necrotizing/diagnosis , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/epidemiology , Female , HMGB1 Protein/metabolism , Incidence , Injections, Subcutaneous , Interleukin-6/metabolism , Male , Pregnancy , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Severity of Illness Index , Solubility , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...