Extravasated platelet aggregation in the livers of rats with drug‑induced hepatic sinusoidal obstruction syndrome

  • Authors:
    • Miki Hirata
    • Hidehiro Tajima
    • Tomoharu Miyashita
    • Takashi Miyata
    • Shinichi Nakanuma
    • Isamu Makino
    • Hironori Hayashi
    • Katsunobu Oyama
    • Hiroyuki Takamura
    • Itasu Ninomiya
    • Sachio Fushida
    • Hiroki Nakata
    • Shoichi Iseki
    • Shinichi Harada
    • Tomohiko Wakayama
    • Tetsuo Ohta
  • View Affiliations

  • Published online on: March 28, 2017     https://doi.org/10.3892/mmr.2017.6407
  • Pages: 3147-3152
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Oxaliplatin-based chemotherapy plays an important role in the treatment of colorectal liver metastases. Oxaliplatin, however, causes sinusoidal obstruction syndrome (SOS), which is characterized by portal hypertension, splenomegaly, thrombocytopenia, and liver dysfunction. SOS is diagnosed histopathologically by disruption of the sinusoidal endothelium, collagen deposition, fibrosis especially around zone 3, dilatation of the sinusoidal space and congestion. This study assessed the characteristics of a rat model of SOS. SOS was induced in rats by administration of monocrotaline (MCT). Blood chemistries and macroscopic and microscopic findings were compared in rats administered MCT and vehicle (control group). Levels of expression in the liver of CD41, P‑selectin, rat endothelial cell antigen‑1, CD34, and cleaved caspase‑3 were analyzed immunohistochemically. Moreover, livers of these rats were analyzed by electron microscopy. Macroscopically, MCT‑treated rats showed accumulation of bloody ascites and blue liver and were diagnosed with SOS histologically. Serum concentrations of aspartate aminotransferase (P=0.003), alanine aminotransferase (P=0.008), total‑bilirubin (P=0.012), direct‑bilirubin (P=0.007), indirect‑bilirubin (P=0.003), lactate dehydrogenase (P<0.001) and hyaluronic acid (P=0.016) were significantly higher, and platelet counts significantly lower (P=0.004), in MCT‑treated than in control rats. The livers of MCT‑treated rats were immunohistochemically positive for CD41 and P‑selectin, suggesting platelet aggregates; for rat endothelial cell antigen‑1 and CD34, suggesting sinusoidal endothelial disorder; and for cleaved caspase‑3, suggesting hepatocyte apoptosis. Electron microscopic findings revealed platelet aggregation in the space of Disse in the MCT group. Extravasated platelet aggregation in Disse's space may be involved in the development of SOS.

Introduction

Systemic neoadjuvant chemotherapy can precede conversion surgery, resulting in R0 resection, even in patients with initially unresectable colorectal liver metastases (CRLM). Although oxaliplatin-based chemotherapy plays a central role in the treatment of patients with CRLM, oxaliplatin can induce sinusoidal obstruction syndrome (SOS), which is characterized by hepatic sinusoidal dilatation, hepatocyte atrophy, peri-sinusoidal fibrosis, and nodular regenerative hyperplasia (1). These histological changes have been reported in up to 40% of patients treated with oxaliplatin-based regimens undergoing liver resection (26). SOS prior to major hepatectomy has been associated with increased peri-operative morbidity and prolonged hospital stay (7). Clinically, factors diagnostic of SOS include portal hypertension, splenomegaly, thrombocytopenia, an abnormal indocyanine green (ICG) retention rate and elevated liver enzymes, bilirubin and hyaluronic acid (HA) (812). On pathological examination, SOS is characterized by the disruption of the sinusoidal endothelium, collagen deposition in peri-sinusoidal spaces, fibrosis especially around the central vein (in zone 3), dilatation of the sinusoidal space and congestion (13). Platelets were shown to be involved in the alterations in the sinusoidal endothelium observed in patients with SOS (14). Thrombocytopenia has been observed in patients with CRLM treated with oxaliplatin based chemotherapy, and examination of their resected livers showed platelet aggregation in zone 3 (15). Platelet aggregation in the extra-sinusoidal (Disse's) space is called extravasated platelet aggregation (EPA). This study assessed whether EPA was present in the livers of rats with drug induced SOS.

Materials and methods

Reagents

Monocrotaline (MCT) was purchased from Wako Pure Chemical Industries (Osaka, Japan). A 20 mg/ml solution of MCT was prepared by dissolving 1,000 mg MCT in 1.0 N HCl, and adjusting the pH to 7.4 with 0.5 N NaOH, followed by dilution in phosphate-buffered saline (PBS), pH 7.4, to increase the total volume to 50 ml (16,17).

Animals

Male Wistar rats, weighing 230–300 g, were purchased from Charles River Inc. (Kanagawa, Japan), and allowed free access to water and standard laboratory chow. This study complied with the guidelines of the Division for Animal Research Resources, University of Kanazawa. All experiments and procedures were approved by the Animal Care and Use Committee of the University of Kanazawa.

Experimental protocol

The animals were randomly divided into two groups of 10 rats each. Animals were fasted for 12 h, with one group administered MCT (90 mg/kg) and the control group administered water (Fig. 1). Rats were thereafter allowed free access to water and standard laboratory chow ad libitum. Because histopathological changes 48 h after MCT administration are similar to those in patients with SOS, (16,18) the rats were anesthetized by inhalation of diethyl ether and sacrificed. Blood was collected from the inferior vena cava, and liver tissues were obtained.

Macroscopic examination

The abdominal cavity of rats was accessed by middle incision laparotomy, and the effect of MCT assessed macroscopically by the accumulation of peritoneal fluid and the color of the liver surface.

Biochemical analysis

Blood specimens were collected, and white blood cell (WBC) and platelet counts, as well as hemoglobin (Hb) concentrations, were measured using an automated blood cell counter (Celltac α, MEK-6458, Nihon Kohden, Japan). Serum concentrations of aspartate aminotransferase (AST), alanine aminotransferase (ALT), total bilirubin (T-Bil), direct bilirubin (D-Bil), indirect bilirubin (I-Bil) lactate dehydrogenase (LDH) and hyaluronic acid (HA) were measured by SRL Inc. (Tokyo, Japan).

Histologic analysis

Liver tissue was fixed in 10% neutral buffered formalin, embedded in paraffin, and cut serially into 4-µm sections. Slides were stained with hematoxylin and eosin (H&E), and ten randomly selected high-power fields (magnification, ×200) were examined. The degree of SOS was assessed by examining histological changes in sinusoidal dilatation, coagulative necrosis of hepatocytes, endothelial damage to the central vein, and sinusoidal hemorrhage (16,18,19). Each of these four features was graded on a 4-point scale, with 0, absent; 1, mild (1–30%); 2, moderate (31–60%); and 3, severe (61–100%). The total SOS score for each rat was calculated as the sum of the individual scores.

Immunohistochemistry

Tissue samples were fixed in 4% paraformaldehyde in phosphate buffered saline for 3 days and embedded in a solution of O.C.T. compound (Sakura Finetek, Tokyo, Japan) and 30% sucrose in 0.1 M phosphate buffer, pH 7.4, containing 0.05% NaN3. All tissue samples were sectioned at 6 µm using cryostats (Thermo Fisher Scientific, Waltham, MA, USA).

Immunohistochemical staining was performed with the Dako Envision system (Dako, Carpinteria, CA, USA), which uses dextran polymers conjugated with horseradish peroxidase, thus avoiding contamination by endogenous biotin. Tissue samples were dissected out, immersed in fixative for 4 h, dehydrated in a graded alcohol series for 24 h, embedded in paraffin, and cut into 4-mm-thick sections. Endogenous peroxidases were blocked by immersing the sections in 3% H2O2 in 100% methanol for 20 min at room temperature. Antigen retrieval was performed by micro-waving sections at 95°C for 10 min in 0.001 M citrate buffer (pH 6.7). The sections were incubated with serum-free protein block (Dako) at room temperature for 10 min to block non-specific staining, followed by incubation for 2 h at room temperature with primary monoclonal antibody. Peroxidase activity was detected with the enzyme substrate 3-amino-9-ethyl carbazole. As negative controls, tissue sections were incubated with Tris-buffered saline without the primary antibodies. All samples were counterstained with Meyer's hematoxylin.

Platelet aggregation was assessed by immunohistochemical staining of liver tissues with antibodies to platelet glycoprotein IIb (CD41) (1:100, orb4832; Biorbyt, Cambridge, UK) and P-selectin (1:50, ERP1444(2)(B); Abcam, Tokyo, Japan), the latter being a member of the selectin family of adhesion molecules expressed on activated platelets and endothelium (20). Damage to sinusoidal endothelial cells was determined by immunostaining rat liver tissues with antibodies to rat endothelial cell antigen-1 (RECA-1) (#MCZ-970R; Serotec, Oxford, UK) and CD34 (1:20, AF4117; R&D Systems, Inc., Minneapolis, MN, USA), the latter being a marker of sinusoidal capillary formation in liver tissue, thereby differentiating between normal and altered sinusoidal epithelium (2124). Hepatocyte apoptosis was determined by immunostaining with antibody to cleaved caspase-3 (1:100, 9661; Cell Signaling Technology, Inc., Beverly, MA, USA). Dyeabilities of these factors were measured with the BZ-Analyzer software (Keyence, Osaka, Japan). The total stained areas were calculated from randomly selected images per high power field (magnification, ×200) and compared between the two groups.

Transmission electron microscopy

Rat liver specimens were fixed by immersion in 2% paraformaldehyde plus 2.5% glutaraldehyde in phosphate buffer (pH 7.4) for 4 h at 4°C. After washing in phosphate buffer at 4°C, the specimens were postfixed in 1% osmium tetraoxide for 2 h at 4°C, washed repeatedly in distilled water, stained with 1% uranyl acetate for 30 min, dehydrated through a graded ethanol series and propylene oxide, and embedded in Glicidether (Selva Feinbiochemica, Heidelberg, Germany). Ultrathin sections were cut and mounted onto copper grids, stained with 1% uranyl acetate for 10 min and with Reynolds lead citrate for 5 min, and evaluated using an H7650 electron microscope (Hitachi, Tokyo, Japan) (25).

Statistical analysis

All results were expressed as the mean ± standard deviation (SD). Groups were compared by Student's t-tests. P<0.05 was considered to indicate a statistically significant difference.

Results

At sacrifice, macroscopic examination of the livers in the MCT group showed accumulation of bloody ascites, with the liver surface appearing congested and dark red in color (Fig. 1). These changes were not observed in the control group. The difference of liver size and weight was not observed between the two groups.

The results of complete blood counts and serum biochemistry tests are shown in Table I. There were no between group differences in WBC count and Hb concentration, although platelet counts were significantly lower in the MCT than in the control group (P=0.004). Serum concentrations of AST (P=0.003), ALT (P=0.008), T-Bil (P=0.012), D-Bil (P=0.007), I-Bil (P=0.003), LDH (P<0.001) and HA (P=0.0016) were all significantly higher in the MCT than in the control group.

Table I.

Blood counts and serum biochemistry.

Table I.

Blood counts and serum biochemistry.

VariablesMCT group (n=10)Control group (n=10)P-value
Hb (g/dl)14.73±1.4314.34±1.040.281
WBC (x103/µl)48.6±17.873.6±11.10.232
Plt (x103/µl)5.87±2.775.78±8.30.004
AST (IU/l)7,218.0±4071.382.9±9.10.003
ALT (IU/l)1,539.2±837.733.4±5.40.008
T-Bil (mg/dl)0.211±0.0830.038±0.0070.012
D-Bil (mg/dl)0.126±0.0530.032±0.0040.007
I-Bil (mg/dl)0.085±0.0380.006±0.0050.003
LDH (mg/dl) 5,664.9±3,700.9312.9±158.7<0.001
HA (ng/dl)1,188.3±729.036.8±3.90.016

[i] Hb, hemoglobin; WBC, white blood cell; Plt, platelet; AST, spartate aminotransferase; ALT, alanine aminotransferase; T-Bil, total bilirubin; D-Bil, direct bilirubin; I-Bil, indirect bilirubin; LDH, lactate dehydrogenase; HA, hyaluronic acid.

Histologically, H&E staining of liver tissue showed higher SOS scores in the MCT than in the control group, with the former group showing all four histological changes: sinusoidal dilatation, coagulative necrosis of hepatocytes, endothelial damage to the central vein, and sinusoidal hemorrhage (Fig. 2).

CD41 protein expression was observed in the MCT group, especially in zone 3, but not in the control group (Fig. 3). Assessment of P-selectin expression showed that, in the control group, it was present only on the endothelial cells of the sinusoids (Fig. 4A). In the MCT group, however, P-selectin expression was observed on both sinusoidal endothelial cells and granularly stained platelets (Fig. 4B).

RECA-1 protein expression was markedly lower in the MCT (Fig. 5B) than in the control group (Fig. 5A), indicating that the sinusoidal lining had largely disappeared from the former. In contrast, CD34 expression was higher in the MCT group, especially in zone 3 (Fig. 6B), than in the control group (Fig. 6A). Similarly, cells positive for cleaved caspase-3 were observed in zone 3 in the MCT group 3 (Fig. 7B), but not in the control group. Measurements of the staining areas of MCT and control group are shown in Table II. In the MCT group, significant excessive staining was observed in all the factors in comparison with the control group.

Table II.

Expression areas of immunohistochemical examination

Table II.

Expression areas of immunohistochemical examination

VariablesMCT group (µm2)Control group (µm2)P-value
CD41 2,405.0±2,476.3760.0±11.80.014
P-selectin 14,653.7±2,324.011,678.1±505.00.034
RECA-151,503.0±174.0 246,664.0±64,171.00.001
CD34113,914±5,33618,720±4,780<0.001
Cleaved974±3358.3±0.70.003
Caspase-3

[i] RECA-1, rat endothelial cell antigen-1.

Electron microscopic examination of the MCT group showed many platelets containing granules in the space of Disse (Fig. 8).

Discussion

SOS is a veno-occlusive disease (VOD), characterized by hepatic injury after exposure to a drug or toxin or after bone marrow transplantation (26). SOS can present in acute, subacute and chronic forms, usually as abdominal pain and/or swelling, with evidence of portal hypertension and variable degrees of serum enzyme elevation and jaundice. Liver histology shows obstruction of the sinusoids in the central areas, along with hepatocyte necrosis and hemorrhage. Agents that cause SOS include cancer chemotherapeutic agents, particularly alkylating agents such as cyclophosphamide and the platinum coordination complexes cisplatin and oxaliplatin. The diagnosis of SOS usually depends on a typical clinical presentation, characterized by hepatomegaly, ascites, portal hypertension, weight gain and jaundice (27), or by the exclusion of other causes of liver injury. The usual differential diagnosis of SOS after bone marrow transplantation includes graft vs. host disease, sepsis, other forms of drug induced liver injury, and various forms of viral hepatitis. SOS diagnosis is usually supported by imaging modalities that demonstrate changes typical of sinusoidal obstruction. Liver biopsy is diagnostic but is not usually needed; moreover, concurrent coagulopathy or thrombocytopenia make it difficult to acquire biopsy samples (28). Although thrombocytopenia in SOS usually results from hypersplenism, decreased platelet counts have been observed earlier than splenomegaly (15). Therefore, this research focused on the role of platelets in the pathogenesis of SOS.

Histologic examination of the liver in patients with SOS shows obstruction of the sinusoids around the central vein (zone 3), along with hepatocyte necrosis and congestion. SOS may be caused by cancer chemotherapeutic and immunosuppressing agents. SOS was shown to be induced by oxaliplatin based chemotherapy in patients with CRLM, appearing as blue liver. Administration of MCT to rats induced SOS, with the macroscopic appearance of blue liver and SOS detected histologically. Blood analysis showed that treatment with MCT significantly increased serum concentrations of AST, ALT, T-Bil, D-Bil and HA, with the latter reflecting injury to liver sinusoidal endothelial cells (LSECs) (13).

Histopathologically, administration of MCT resulted in sinusoidal dilatation, coagulative necrosis of hepatocytes, endothelial damage to the central vein, and sinusoidal hemorrhage. Immunohistochemical assays showed that MCT administration resulted in reduced RECA-1 and increased CD34 expression. Reduced RECA-1 expression indicated that the sinusoidal lining had largely disappeared. CD34 has been used to detect sinusoidal capillary formation in liver tissue and to differentiate between normal and altered sinusoidal epithelium (2023).

Neutrophils and platelets participate in the pathogenesis of severe sepsis, as well as forming neutrophil extracellular trapping systems (NETs) to kill pathogens extracellularly (29). NETs, however, have been reported to contribute to organ damage in patients with infectious diseases (30) Furthermore, sivelestat sodium, a neutrophil elastase inhibitor, was found to inhibit neutrophil adhesion and migration to vascular endothelium during hepatic ischemia reperfusion and prevent liver injury (31). Platelets are not stained by H&E. because they lack nuclei. Platelets can be visualized in rats by immunostaining with antibody to the surface marker, CD41. Clinically, we observed EPA in SOS patients after liver transplantation (32,33) and after oxaliplatin chemotherapy (15). Platelets can be indirectly localized by detection of their surface markers. However, it was possible to directly verify the presence of platelets in the space of Disse around zone 3 by electron microscopy in the MCT group.

SOS has also been referred to as VOD in transplantation. Autopsy reports have shown that the pathology of human VOD includes reticulin deposition within sinusoids, central vein occlusion, hepatocyte atrophy/necrosis, sinusoidal hemorrhage, and sparing of portal tracts (34). Earlier histopathological findings of VOD can be detected by examining liver biopsy specimens. The affected venules show a lumen narrowed by a wide, edematous subendothelial zone containing a fibrous material and fragmented red cells. The sinusoids draining into these veins are congested, and the surrounding hepatocytes are pale and necrotic. Hemorrhage is also observed in the space of Disse.

Although sinusoidal fibrosis in zone 3 is regarded as an equivalent of early VOD, this diagnosis is not usually accepted unless there are significant changes in the central vein (35). Despite venous reflux disorder, with portal blood flowing back, being observed in patients with severe SOS, but venous obstruction or thrombosis has not been detected pathologically. Platelets not only form a white thrombus, but may cause spasms in the central vein by secreting various growth factors, including thromboxane A2 (TXA2), vascular endothelial growth factor (VEGF)-A, transforming growth factor (TGF)-β and plasminogen activator inhibitor (PAI)-1 (36). These growth factors can explain the liver injury in SOS. For example, TXA2 can cause central vein occlusion and portal hypertension, (37) and thrombospondin-activated TGF-β can cause collagen deposition in the peri-sinusoidal space and inhibit substance exchange in the space of Disse (28). TGF-β can also increase serum bilirubin concentration and may be responsible for an abnormal ICG retention rate. PAI-1 and TGF-β interfere with liver regeneration by suppressing hepatocyte growth factor (38). Although VEGF-A usually acts as a vasodilator, it can act as a vasoconstrictor under conditions of endothelial failure and hepatic sinusoidal injury (39). Platelet aggregation in the space of Disse may explain many of the symptoms, venous perfusion abnormalities, and portal hypertension detected in SOS, findings that cannot be confirmed pathologically. Thus, antiplatelet agents, including vasodilators such as prostaglandin E1 (40) and low dose heparin, (41) may prevent or reduce SOS.

In conclusion, EPA in the space of Disse has been confirmed in this rat model of MCT-induced liver SOS. Platelets are heavily involved in the development of SOS.

References

1 

Rubbia-Barandt L, Audard V, Sartoretti P, Roth AD, Brezault C, Le Charpentier M, Dousset B, Morel P, Soubrane O, Chaussade S, et al: Severe hepatic sinusoidal obstruction associated with oxaliplatin-based chemotherapy in patients with metastatic colorectal cancer. Ann Oncol. 15:460–466. 2004. View Article : Google Scholar : PubMed/NCBI

2 

Hubert C, Fevaille C, Sempoux C, Horsmans Y, Humblet Y, Machiels JP, Zech F, Ceratti A and Gigot JF: Prevalence and clinical relevance of pathological hepatic changes occurring after neoadjuvant chemotherapy for colorectal liver metastases. Surgery. 147:185–194. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Vauthey JN, Pawlik TM, Ribero D, Wu TT, Zorzi D, Hoff PM, Xiong HQ, Eng C, Lauwers GY, Mino-Kenudson M, et al: Chemotherapy regimen predicts steatohepatitis and an increase in 90-day mortality after surgery for hepatic colorectal metastases. J Clin Oncol. 24:2065–2072. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Tamandl D, Klinger M, Eipeldauer S, Herberger B, Kaczirek K, Gruenberger B and Gruenberger T: Sinusoid obstruction syndrome impairs long-term outcome of colorectal liver metastases treated with resection after neoadjuvant chemotherapy. Ann Surg Oncol. 18:421–430. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Robinson SM, Wilson CH, Burt AD, Manas DM and White SA: Chemotherapy associated liver injury in patients with colorectal liver metastases: A systemic review and meta-analysis. Ann Surg Oncol. 19:4287–4299. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Aloysius MM, Zaitoun AM, Beckingham IJ, Neal KR, Aithal GP, Bessell EM and Lobo DN: The pathological response to neoadjuvant chemotherapy with FOLFOX-4 for colorectal liver metastases: A comparative study. Virchows Arch. 451:943–948. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Nakano H, Oussoultzoqlou E, Rosso E, Casnedi S, Chenard-Neu MP, Dufour P, Bachellier P and Jaeck D: Sinusoidal injury increases morbidity after major hepatectomy in patients with colorectal liver metastases receiving preoperative chemotherapy. Ann Surg. 247:118–124. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Jardim DL, Rodorigues CA, Novis YA, Rocha VG and Hoff PM: Oxaliplatin-related thrombocytopenia. Ann Oncol. 23:1937–1942. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Miura K, Nakano H, Sakurai J, Kobayashi S, Koizumi S, Arai T, Shimamura T, Makizumi R, Yamada K, Miyajima N, et al: Splenomegaly in FOLFOX-naïve stage IV or recurrent colorectal cancer patients due to chemotherapy- associated hepatotoxicity can be predicted by the aspartate aminotransferase to platelet ratio before chemotherapy. Int J Clin Oncol. 16:257–263. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Klieger PM, Tamandl D, Herberger B, Faybik P, Fleichmann E, Maresch J and Gruengerger T: Evaluation of chemotherapy-associated liver injury in patients with colorectal cancer liver metastases using indocyanine green clearance testing. Ann Surg Oncol. 18:1644–1650. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Sato S, Nakano H, Ishida Y and Otsubo T: The aspartate aminotransferase to platelet ratio before chemotherapy predicts adverse events for FOLFOX and XELOX regimens including bevacizumab as the first-line therapy for stage IV, recurrent and metastatic colorectal cancer. J Gastrointest Oncol. 4:203–209. 2013.PubMed/NCBI

12 

Narita M, Oussoultzoglou E, Chenard MP, Fuchshuber P, Rather M, Rosso E, Addeo P, Jaeck D and Bachellier P: Liver injury due to chemotherapy-induced sinusoidal obstruction syndrome is associated with sinusoidal capillarization. Ann Surg Oncol. 19:2230–2237. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Morine Y, Shimada M and Utsunomiya T: Evaluation and management of hepatic injury induced by oxaliplatin-based chemotherapy in patients with hepatic resection for colorectal liver metastasis. Hepatol Res. 44:59–69. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Lalor PF, Herbert J, Bicknell R and Adams DH: Hepatic sinusoidal endothelium avidly binds platelets in an integrin-dependent manner, leading to platelet and endothelial activation and leucocyte recruitment. Am J Physiol Gastrointest Liver Physiol. 304:G469–G478. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Tajima H, Ohta T, Miyashita T, Nakanuma S, Matoba M, Miyata T, Sakai S, Okamoto K, Makino I, Kinoshita J, et al: Oxaliplatin-based chemotherapy induces extravasated platelet aggregation in the liver. Mol Clinic Oncol. 3:555–558. 2015.

16 

Narita M, Hatano E, Ikai I, Miyagawa-Hayashino A, Yanagida A, Nagata H, Asechi H, Taura K and Uemoto S: A phosphodiesterase III inhibitor protects rat liver from sinusoidal obstruction syndrome through heme oxygenase-1 induction. Ann Surg. 249:806–813. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Prié S, Stewart DJ and Dupuis J: EndothelinA receptor blockade improves nitric oxide-mediated vasodilation in monocrotaline-induced pulmonary hypertension. Circulation. 97:2169–2174. 1998. View Article : Google Scholar : PubMed/NCBI

18 

Nakamura K, Hatano E, Narita M, Miyagawa-Hayashino A, Koyama Y, Nagata H, Iwaisako K, Taura K and Uemoto S: Sorafenib attenuates monocrotaline-induced sinusoidal obstruction syndrome in rats through suppression of JNK and MMP-9. Hepatology. 57:1037–1043. 2012. View Article : Google Scholar

19 

DeLeve LD, McCuskey RS, Wang X, Hu L, McCuskey MK, Epstein RB and Kanel GC: Characterization of a reproducible rat model of hepatic veno-occlusive disease. Hepatology. 29:1779–1791. 1999. View Article : Google Scholar : PubMed/NCBI

20 

Suzuki M, Bachelet-Violette L, Rouzet F, Beilvert A, Autret G, Maire M, Menager C, Louedec L, Choqueux C, Saboural P, et al: Ultrasmall superparamagnetic iron oxide nanoparticles coated with fucoidan for molecular MRI of intraluminal thrombus. Nanomedicine (Lond). 10:73–87. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Wisse E, De Zanger RB, Charels K, Van Der Smissen P and McCuskey RS: The liver sieve: Considerations concerning the structure and function of endothelial fenestrae, the sinusoidal wall and the space of Disse. Hepatology. 5:683–692. 1985. View Article : Google Scholar : PubMed/NCBI

22 

Fina L, Molgaard HV, Robertson D, Bradley NJ, Monaghan P, Delia D, Sutherland DR, Baker MA and Greaves MF: Expression of the CD34 gene in vascular endothelial cells. Blood. 75:2417–2426. 1990.PubMed/NCBI

23 

Couvelard A, Scoazec JY, Dauge MC, Bringuier AF, Potet F and Feldmann G: Structural and functional differentiation of sinusoidal endothelial cells during liver organogenesis in humans. Blood. 87:4568–4580. 1996.PubMed/NCBI

24 

Kuntz E and Kuntz HD: Textbook and Atlas: History, Morphology, Biochemistry, Diagnostics, Clinic, TherapyHepatology. 3rd. Springer Medizin Verlag; Heidelberg: 2008, View Article : Google Scholar

25 

Wakayama T, Nakata H, Kurobo M, Sai Y and Iseki S: Expression, localization, and binding activity of the ezrin/radixin/moesin proteins in the mouse testis. J Histochem Cytochem. 57:351–362. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Jelliffe DB, Bras G and Stuart KL: Veno-occlusive disease of the liver. Pediatrics. 14:334–339. 1954.PubMed/NCBI

27 

Campos-Varela I, Castells L, Dopazo C, Pérez-Lafuente M, Allende H, Len O, Llopart L, Vargas V and Charco R: Transjuglar intrahepatic portsystemic shunt for the treatment of sinusoidal obstruction syndrome in a liver transplant recipient and review of the literature. Liver Transpl. 18:201–205. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Yao L, Yao ZM and Yu T: Influence of BOL on hyaluronic acid, laminin and hyperplasia in hepatofibrotic rats. World J Gastroenterol. 7:872–875. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y and Zychlinsky A: Neutrophil extracellular traps kill bacteria. Science. 303:1532–1535. 2001. View Article : Google Scholar

30 

Merza M, Hartman H, Rahman M, Hwaiz R, Zhang E, Renström E, Luo L, Mörgelin M, Regner S and Thorlacius H: Neutrophil extracellular traps induce trypsin activation, inflammation and tissue damage in mice with severe acute pancreatitis. Gastroenterology. 149:1920–1931, e8. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Sakai S, Tajima H, Miyashita T, Nakanuma S, Makino I, Hayashi H, Nakagawara H, Kitagawa H, Fushida S, Fujimura T, et al: Sivelestat sodium hydrate inhibits neutrophil migration to the vessel wall and suppresses hepatic ischemia reperfusion injury. Dig Dig Sci. 59:787–794. 2014. View Article : Google Scholar

32 

Nakanuma S, Miyashita T, Hayashi H, Tajima H, Takamura H, Tsukada T, Okamoto K, Sakai S, Makino I, Kinoshita J, et al: Extravasated platelet aggregation in liver zone 3 may correlate with the progression of sinusoidal obstruction syndrome following living donor liver transplantation: A case report. Exp Therap Med. 9:1119–1124. 2015.

33 

Takamura H, Nakanuma S, Hayashi H, Tajima H, Kakinoki K, Kitahara M, Sakai S, Makino I, Nakagawara H, Miyashita T, et al: Severe veno-occlusive disease/sinusoidal obstruction syndrome after deceased-donor and living-donor liver transplantation. Transplant Proc. 46:3523–3535. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Pai RK, van Besien K, Hart J, Artz AS and O'Donnell PH: Clinicopathologic features of late onset veno-occlusive disease/sinusoidal obstructive syndrome after high dose busulfan and hematopoietic cell transplantation. Leuk Lymphoma. 53:1552–1557. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Carreras E, Grañena A and Rozman C: Hepatic veno-occlusive disease after bone marrow transplant. Blood Rev. 7:43–51. 1993. View Article : Google Scholar : PubMed/NCBI

36 

Battinelli EM, Markens BA and Italiano JE Jr: Release of angiogenesis regulatory proteins from platelet alpha granules: Modulation of physiologic and pathologic angiogenesis. Blood. 118:1359–1368. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Cui S, Shibamoto T, Liu W, Takano H and Kurata Y: Effects of platelet- activating factor, thromboxane A2 and leukotriene D4 on isolated perfused rat liver. Prostaglandins Other Lipid Mediat. 80:35–45. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Narmada BC, Chia SM, Tucker-Kellogg L and Yu H: HGF regulates the activation on TGF-β1 in rat hepatocytes and hepatic stellate cells. J Cell Physiol. 228:393–401. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Parenti A, Brogelli L, Filippi S, Donnini S and Ledda F: Effect of hypoxia and endothelial loss on vascular smooth muscle cell responsiveness to VEGF-A: Role of fit-1/VEGF-receptor-1. Cardiovasc Res. 55:201–212. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Oates JA, FitzGerald GA, Branch RA, Jackson EK, Knapp HR and Roberts LJ II: Clinical implications of prostaglandin and thromboxane A2 formation (1). N Eng J Med. 319:689–698. 1988. View Article : Google Scholar

41 

Attal M, Huguet F, Rubie H, Huynh A, Charlet JP, Payen JL, Voigt JJ, Brousset P, Selves J and Muller C: Prevention of hepatic veno-occlusive disease after bone marrow transplantation by continuous infusion of low-dose heparin: A prospective, randomized trial. Blood. 79:2834–2840. 1992.PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 15 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hirata M, Tajima H, Miyashita T, Miyata T, Nakanuma S, Makino I, Hayashi H, Oyama K, Takamura H, Ninomiya I, Ninomiya I, et al: Extravasated platelet aggregation in the livers of rats with drug‑induced hepatic sinusoidal obstruction syndrome. Mol Med Rep 15: 3147-3152, 2017
APA
Hirata, M., Tajima, H., Miyashita, T., Miyata, T., Nakanuma, S., Makino, I. ... Ohta, T. (2017). Extravasated platelet aggregation in the livers of rats with drug‑induced hepatic sinusoidal obstruction syndrome. Molecular Medicine Reports, 15, 3147-3152. https://doi.org/10.3892/mmr.2017.6407
MLA
Hirata, M., Tajima, H., Miyashita, T., Miyata, T., Nakanuma, S., Makino, I., Hayashi, H., Oyama, K., Takamura, H., Ninomiya, I., Fushida, S., Nakata, H., Iseki, S., Harada, S., Wakayama, T., Ohta, T."Extravasated platelet aggregation in the livers of rats with drug‑induced hepatic sinusoidal obstruction syndrome". Molecular Medicine Reports 15.5 (2017): 3147-3152.
Chicago
Hirata, M., Tajima, H., Miyashita, T., Miyata, T., Nakanuma, S., Makino, I., Hayashi, H., Oyama, K., Takamura, H., Ninomiya, I., Fushida, S., Nakata, H., Iseki, S., Harada, S., Wakayama, T., Ohta, T."Extravasated platelet aggregation in the livers of rats with drug‑induced hepatic sinusoidal obstruction syndrome". Molecular Medicine Reports 15, no. 5 (2017): 3147-3152. https://doi.org/10.3892/mmr.2017.6407