Response: Estimating frequency of cancer stem cells in a mouse model of adult T-cell leukemia/lymphoma

Blood ◽  
2010 ◽  
Vol 115 (10) ◽  
pp. 2118-2118
Author(s):  
Jumpei Yamazaki ◽  
Takuo Mizukami ◽  
William W. Hall ◽  
Isao Hamaguchi
Retrovirology ◽  
2011 ◽  
Vol 8 (Suppl 1) ◽  
pp. A9 ◽  
Author(s):  
Tadaki Suzuki ◽  
Akiko Okayama ◽  
Takahiro Tsuji ◽  
Akihide Ryo ◽  
Hisashi Hirano ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (13) ◽  
pp. 2709-2720 ◽  
Author(s):  
Jumpei Yamazaki ◽  
Takuo Mizukami ◽  
Kazuya Takizawa ◽  
Madoka Kuramitsu ◽  
Haruka Momose ◽  
...  

AbstractAdult T-cell leukemia/lymphoma (ATL) is a malignant lymphoproliferative disorder caused by HTLV-I infection. In ATL, chemotherapeutic responses are generally poor, which has suggested the existence of chemotherapy-resistant cancer stem cells (CSCs). To identify CSC candidates in ATL, we have focused on a Tax transgenic mouse (Tax-Tg) model, which reproduces ATL-like disease both in Tax-Tg animals and also after transfer of Tax-Tg splenic lymphomatous cells (SLCs) to nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. Using a limiting dilution transplantation, it was estimated that one CSC existed per 104 SLCs (0.01%). In agreement with this, we have successfully identified candidate CSCs in a side population (0.06%), which overlapped with a minor population of CD38−/CD71−/CD117+ cells (0.03%). Whereas lymphoma did not develop after transplantation of 102 SLCs, 102 CSCs could consistently regenerate the original lymphoma. In addition, lymphoma and CSCs could also be demonstrated in the bone marrow and CD117+ CSCs were observed in both osteoblastic and vascular niches. In the CSCs, Tax, Notch1, and Bmi1 expression was down-regulated, suggesting that the CSCs were derived from Pro-T cells or early hematopoietic progenitor cells. Taken together, our data demonstrate that CSCs certainly exist and have the potential to regenerate lymphoma in our mouse model.


2019 ◽  
Vol 19 ◽  
pp. 100257 ◽  
Author(s):  
Nicole A. Kohart ◽  
Said M. Elshafae ◽  
Wachirapan Supsahvad ◽  
Aylin Alasonyalilar-Demirer ◽  
Amanda R. Panfil ◽  
...  

2007 ◽  
Vol 67 (24) ◽  
pp. 11859-11866 ◽  
Author(s):  
S. T. Shu ◽  
M. V.P. Nadella ◽  
W. P. Dirksen ◽  
S. A. Fernandez ◽  
N. K. Thudi ◽  
...  

2005 ◽  
Vol 26 (8) ◽  
pp. 1382-1388 ◽  
Author(s):  
Takeo Ohsugi ◽  
Ryouichi Horie ◽  
Toshio Kumasaka ◽  
Akira Ishida ◽  
Takaomi Ishida ◽  
...  

PLoS ONE ◽  
2021 ◽  
Vol 16 (8) ◽  
pp. e0256320
Author(s):  
Yanuar Rahmat Fauzi ◽  
Shingo Nakahata ◽  
Syahrul Chilmi ◽  
Tomonaga Ichikawa ◽  
Phawut Nueangphuet ◽  
...  

Adult T-cell leukemia/lymphoma (ATLL) originates from human T-cell leukemia virus type 1 (HTLV-1) infection due to the activation of the nuclear factor-κB (NF-κB) signaling pathway to maintain proliferation and survival. An important mechanism of the activated NF-κB signaling pathway in ATLL is the activation of the macroautophagy (herafter referred to as autophagy in the remainder of this manuscript)-lysosomal degradation of p47 (NSFL1C), a negative regulator of the NF-κB pathway. Therefore, we considered the use of chloroquine (CQ) or hydroxychloroquine (HCQ) (CQ/HCQ) as an autophagy inhibitor to treat ATLL; these drugs were originally approved by the FDA as antimalarial drugs and have recently been used to treat autoimmune diseases, such as systemic lupus erythematosus (SLE). In this paper, we determined the therapeutic efficacy of CQ/HCQ, as NF-κB inhibitors, in ATLL mediated by blockade of p47 degradation. Administration of CQ/HCQ to ATLL cell lines and primary ATLL cells induced cell growth inhibition in a dose-dependent manner, and the majority of cells underwent apoptosis after CQ administration. As to the molecular mechanism, autophagy was inhibited in CQ-treated ATLL cells, and activation of the NF-κB pathway was suppressed with the restoration of the p47 level. When the antitumor effect of CQ/HCQ was examined using immunodeficient mice transplanted with ATLL cell lines, CQ/HCQ significantly suppressed tumor growth and improved the survival rate in the ATLL xenograft mouse model. Importantly, HCQ selectively induced ATLL cell death in the ATLL xenograft mouse model at the dose used to treat SLE. Taken together, our results suggest that the inhibition of autophagy by CQ/HCQ may become a novel and effective strategy for the treatment of ATLL.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1425-1425
Author(s):  
Prabal Banerjee ◽  
Michael D Lairmore ◽  
Juan Carlos Ramos ◽  
William J Harrington ◽  
Mark A Beilke ◽  
...  

Abstract Abstract 1425 Poster Board I-448 Human T-lymphotropic virus type-1 (HTLV-1) is the first human retrovirus linked to cancer and is the etiologic agent of adult T-cell leukemia/lymphoma (ATLL), an aggressive CD4+ T cell malignancy. The molecular and genetic factors induced by HTLV-1 that initiate ATLL remain unclear, in part, due to the lack of an animal model which recapitulates leukemogenic events. In particular, early target cell infection and transformation events have not been identified or defined. Herein, we have created humanized NOD/SCID (HU-NOD/SCID) mice by inoculation of NOD/SCID mice with CD34+ hematopoietic progenitor and stem cells (CD34+ HP/HSCs) infected ex vivo with HTLV-1. HTLV-1-HU-NOD/SCID mice consistently developed CD4+ T cell lymphomas with characteristics similar to ATLL. Elevated proliferation of infected human stem cells (CD34+CD38−) in the bone marrow was observed in mice developing malignancies. Furthermore, examination of CD34+ HP/HSCs from HTLV-1-infected patients revealed proviral integrations suggesting a role of human bone marrow-derived stem cells in leukemogenesis. NOD/SCID mice reconstituted with CD34+ HP/HSCs transduced with a lentivirus vector (LV) expressing the HTLV-1 oncoprotein (Tax1) also developed CD4+ lymphomas. The recapitulation of a CD4+ T cell lymphoma in HTLV-1- and Tax1-HU-NOD/SCID mice suggest that hematopoietic stem cells serve as a viral reservoir in vivo and provide a cellular target for cell transformation in humans. This animal model of HTLV-1 induced ATLL will provide an important tool for the identification of molecular and cellular events that control the initiation and progression of the lymphoma and potential therapeutic targets to block tumor development. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 125 (23) ◽  
pp. 3527-3535 ◽  
Author(s):  
Yuya Nagai ◽  
Masahiro Kawahara ◽  
Masakatsu Hishizawa ◽  
Yayoi Shimazu ◽  
Noriko Sugino ◽  
...  

Key Points ATL clones are preserved in a rare CD4+CD45RA+ TSCM population. ATL-TSCM cells unidirectionally produce conventional CD45RO+ ATL cells and show a high potency of repopulating identical ATL clones in vivo.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3691-3691
Author(s):  
Takuo Mizukami ◽  
Kazuya Takizawa ◽  
Jumpei Yamazaki ◽  
Wakako Kuribayashi ◽  
Madoka Kuramitsu ◽  
...  

Abstract Adult T cell leukemia (ATL) is a lymphoproliferative disorder caused by infection with HTLV-I. Although various chemotherapies have shown significant complete remission rates, most of the treated patients relapse. These data indicate the existence of leukemic stem cells (LSCs) and a specific niche that regulates stemness and protects LSCs from various chemotherapies. We have reported in previous studies that the ATL-LSCs isolated from a Tax-transgenic (Tax-Tg) mouse are enriched in the CD117+/CD38–/CD71– fraction of the lymphoma, and LSCs have the potential to reproduce the original tumor when transplanted into a NOD/SCID mouse (Yamazaki et al., Blood, 2009). However, the niche of ATL-LSCs is still unclear. To identify the ATL-LSC niche in vivo, we performed a homing assay. Splenic lymphoma cells isolated from a Tax-Tg mouse were GFP transduced by a lentivirus, and then sorted GFP+ cells were transplanted intra-peritoneally into a non-irradiated NOD/SCID mouse. The homing of GFP+ cells to tissues was assessed by flow cytometry (FCM) at 16 hours and 3, 7, 14 and 21 days after transplantation. As a result, GFP+ lymphoma cells were first detected in the spleen and BM at 16 hours after transplantation. No GFP+ lymphoma cells were detected in the thymus and LN. Interestingly, more than 60% of first colonized cells in the spleen and BM at 16 hours after transplantation were AT-LSCs (GFP+/CD117+ cells). From day 3 to 7, more than 40% of colonizing cells in the BM and spleen were ATL-LSCs. To identify the specific niche of ATL-LSCs in the BM, we performed imaging analysis of ATL-LSCs. ATL-LSCs (GFP+/CD117+ and CD38–/CD71–/CD117+ cells) were mainly localized near the endosteal region of trabecular bone in the BM. We found that ATL-LSCs were also attached to the reticular cells in the trabecular bone. In addition, we found the number of osteoclast was significantly increased at the trabecular region. Increasing number of osteoclasts correlates the increased the serum calcium concentration and decreased the mass of trabecular bone. FCM analysis and in vitro differentiation assay confirmed that the number of osteoclast precursors was increased in the ATL BM. To clarify the role of osteoclast in the ATL BM, we treated osteoclast inhibitor Zoledronic acid (ZOL) to the ATL mouse model. As a result, ZOL itself significantly reduced the number of GFP+ ATL cells in the BM. When we treated ZOL with anti cancer drug, GFP+ ATL cells were dramatically reduced in the BM and extend the mouse survival rate significantly despite anti cancer drug does not reduced the number of ATL cells itself. In addition, abnormal trabecular bone morphology was completely recovered in the treated mouse. These data suggest that osteoclast may have a function to support leukemic stem cell niche. To clarify the key signals to induce osteoclast in ATL BM, we checked the expression of RANKL and PTHrP. We found that RANKL was up-regulated both in the lymphoma cell and stromal cells in the bone marrow.   In this study, we found that ATL-LSC niche is located at the trabecular bone region in the BM and osteoclasts have a role to support ATL cell and develop LSCs niche in a mouse model of ATL. We conclude that osteoclast have a potential therapeutic target in the mouse model of ATL. Disclosures: No relevant conflicts of interest to declare.


2013 ◽  
Vol 92 (3) ◽  
pp. 219-228 ◽  
Author(s):  
Fumiko Mori ◽  
Takashi Ishida ◽  
Asahi Ito ◽  
Fumihiko Sato ◽  
Ayako Masaki ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document