scholarly journals In vivo real-time imaging of chemotherapy response on the liver metastatic tumor microenvironment using multiphoton microscopy

2012 ◽  
Vol 28 (5) ◽  
pp. 1822-1830 ◽  
Author(s):  
KOJI TANAKA ◽  
MASATO OKIGAMI ◽  
YUJI TOIYAMA ◽  
YUKI MORIMOTO ◽  
KOHEI MATSUSHITA ◽  
...  
2009 ◽  
Vol 3 (4) ◽  
pp. 351-354 ◽  
Author(s):  
Paul Timpson ◽  
Alan Serrels ◽  
Marta Canel ◽  
Margaret C. Frame ◽  
Valerie G. Brunton ◽  
...  

2013 ◽  
Vol 4 (3) ◽  
pp. 1079 ◽  
Author(s):  
Kehua Xu ◽  
Mingming Qiang ◽  
Wen Gao ◽  
Ruixian Su ◽  
Na Li ◽  
...  

Pharmaceutics ◽  
2021 ◽  
Vol 13 (9) ◽  
pp. 1485
Author(s):  
Aina Venkatasamy ◽  
Eric Guerin ◽  
Anais Blanchet ◽  
Christophe Orvain ◽  
Véronique Devignot ◽  
...  

The reasons behind the poor efficacy of transition metal-based chemotherapies (e.g., cisplatin) or targeted therapies (e.g., histone deacetylase inhibitors, HDACi) on gastric cancer (GC) remain elusive and recent studies suggested that the tumor microenvironment could contribute to the resistance. Hence, our objective was to gain information on the impact of cisplatin and the pan-HDACi SAHA (suberanilohydroxamic acid) on the tumor substructure and microenvironment of GC, by establishing patient-derived xenografts of GC and a combination of ultrasound, immunohistochemistry, and transcriptomics to analyze. The tumors responded partially to SAHA and cisplatin. An ultrasound gave more accurate tumor measures than a caliper. Importantly, an ultrasound allowed a noninvasive real-time access to the tumor substructure, showing differences between cisplatin and SAHA. These differences were confirmed by immunohistochemistry and transcriptomic analyses of the tumor microenvironment, identifying specific cell type signatures and transcription factor activation. For instance, cisplatin induced an “epithelial cell like” signature while SAHA favored a “mesenchymal cell like” one. Altogether, an ultrasound allowed a precise follow-up of the tumor progression while enabling a noninvasive real-time access to the tumor substructure. Combined with transcriptomics, our results underline the different intra-tumoral structural changes caused by both drugs that impact differently on the tumor microenvironment.


2000 ◽  
Vol 113 (20) ◽  
pp. 3663-3671 ◽  
Author(s):  
M. Schrader ◽  
S.J. King ◽  
T.A. Stroh ◽  
T.A. Schroer

We have directly imaged the dynamic behavior of a variety of morphologically different peroxisomal structures in HepG2 and COS-7 cells transfected with a construct encoding GFP bearing the C-terminal peroxisomal targeting signal 1. Real time imaging revealed that moving peroxisomes interacted with each other and were engaged in transient contacts, and at higher magnification, tubular peroxisomes appeared to form a peroxisomal reticulum. Local remodeling of these structures could be observed involving the formation and detachment of tubular processes that interconnected adjacent organelles. Inhibition of cytoplasmic dynein based motility by overexpression of the dynactin subunit, dynamitin (p50), inhibited the movement of peroxisomes in vivo and interfered with the reestablishment of a uniform distribution of peroxisomes after recovery from nocodazole treatment. Isolated peroxisomes moved in vitro along microtubules in the presence of a microtubule motor fraction. Our data reveal that peroxisomal behavior in vivo is significantly more dynamic and interactive than previously thought and suggest a role for the dynein/dynactin motor in peroxisome motility.


2006 ◽  
Author(s):  
Takashi Sakurai ◽  
Seiji Yamamoto ◽  
Atsuo Miyakawa ◽  
Yoshihiko Wakazono ◽  
Takato O. Yoshida ◽  
...  

2016 ◽  
Vol 110 (3) ◽  
pp. 463a
Author(s):  
Fuyu Kobirumaki-Shimozawa ◽  
Kotaro Oyama ◽  
Togo Shimozawa ◽  
Takashi Ohki ◽  
Takako Terui ◽  
...  

Thorax ◽  
2010 ◽  
Vol 65 (Suppl 4) ◽  
pp. A18-A19
Author(s):  
K. Shibuya ◽  
N. Okada ◽  
H. Kohno ◽  
N. Iwai ◽  
M. Noro ◽  
...  

2013 ◽  
Vol 31 (15_suppl) ◽  
pp. e22205-e22205
Author(s):  
Koji Tanaka ◽  
Tadanobu Shimura ◽  
Masato Okigami ◽  
Yuji Toiyama ◽  
Yuhki Morimoto ◽  
...  

e22205 Background: Peritoneal metastasis shows an extremely poor prognosis in patients with gastric cancer. Clinically, the tumor response to chemotherapeutics depends on anatomical location of metastasis. Metastatic tumor xenografts have been shown to be more resistant to chemotherapy than subcutaneous non-metastatic tumor xenografts in preclinical murine model. We have reported a method of in vivo optical pathology using multiphoton microscopy (MPM) in colorectal liver metastatic tumor xenograft model. Aim: We established a method of time-series in vivo optical pathology of peritoneal metastatic xenografts of gastric cancer using MPM. Then, we imaged and evaluated paclitaxel efficacy in the tumor microenvironment with regard to both tumor cell itself and intravascular change in tumor vessels. Methods: Red fluorescent protein (RFP) expressing human gastric cancer cell line (NUGC4) was inoculated into the peritoneal cavity of green fluorescent protein (GFP) expressing nude mice. Paclitaxel (10 mg/kg) was administered three times a week for more than three weeks. Intravital MPM was performed before and after paclitaxel treatment for the exteriorized peritoneal metastatic lesion in the same living mouse. Results: Four to six weeks later, RFP-NUGC4 cells formed macroscopic peritoneal metastases. Red-colored cancer cells and green-colored surrounding stroma with tumor vessels were clearly imaged at the cellular level (in vivooptical pathology). Their cross-sectional images were obtained from the tissue surface to the area of depth of 200 μm (z-stacks imaging). After paclitaxel treatment, tumor cell fragmentation, condensation, swelling and intracellular vacuoles were observed. Within the tumor vessels, platelet aggregation and platelet adhesion to endothelial cells were observed. Conclusions: In vivo optical pathology using MPM provides histopathological information about three-dimensional tissue microarchitecture without tissue shrinkage by fixation and tissue destruction by microtome-sectioning. Our method may become a powerful tool to evaluate the tumor response to new chemotherapeutics on ‘metastatic site’ in preclinical tumor xenograft model.


The Prostate ◽  
2004 ◽  
Vol 59 (4) ◽  
pp. 360-369 ◽  
Author(s):  
Jian Zhang ◽  
Yi Lu ◽  
Jinlu Dai ◽  
Zhi Yao ◽  
Riko Kitazawa ◽  
...  

2011 ◽  
Vol 112 (5) ◽  
pp. 508-510 ◽  
Author(s):  
Kazunori Shimizu ◽  
Yuriko Higuchi ◽  
Yoshimasa Kozu ◽  
Mitsuru Hashida ◽  
Satoshi Konishi

Sign in / Sign up

Export Citation Format

Share Document