Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Linhua Jin is active.

Publication


Featured researches published by Linhua Jin.


Molecular Cancer Therapeutics | 2008

CXCR4 up-regulation by imatinib induces chronic myelogenous leukemia (CML) cell migration to bone marrow stroma and promotes survival of quiescent CML cells

Linhua Jin; Yoko Tabe; Sergej Konoplev; Yuanyuan Xu; Clinton E. Leysath; Hongbo Lu; Shinya Kimura; Akimichi Ohsaka; Mary Beth Rios; Leslie Calvert; Hagop M. Kantarjian; Michael Andreeff; Marina Konopleva

Chronic myelogenous leukemia (CML) is driven by constitutively activated Bcr-Abl tyrosine kinase, which causes the defective adhesion of CML cells to bone marrow stroma. The overexpression of p210Bcr-Abl was reported to down-regulate CXCR4 expression, and this is associated with the cell migration defects in CML. We proposed that tyrosine kinase inhibitors, imatinib or INNO-406, may restore CXCR4 expression and cause the migration of CML cells to bone marrow microenvironment niches, which in turn results in acquisition of stroma-mediated chemoresistance of CML progenitor cells. In KBM5 and K562 cells, imatinib, INNO-406, or IFN-α increased CXCR4 expression and migration. This increase in CXCR4 levels on CML progenitor cells was likewise found in samples from CML patients treated with imatinib or IFN-α. Imatinib induced G0-G1 cell cycle block in CML cells, which was further enhanced in a mesenchymal stem cell (MSC) coculture system. MSC coculture protected KBM-5 cells from imatinib-induced cell death. These antiapoptotic effects were abrogated by the CXCR4 antagonist AMD3465 or by inhibitor of integrin-linked kinase QLT0267. Altogether, these findings suggest that the up-regulation of CXCR4 by imatinib promotes migration of CML cells to bone marrow stroma, causing the G0-G1 cell cycle arrest and hence ensuring the survival of quiescent CML progenitor cells. [Mol Cancer Ther 2008;7(1):48–58]


Cancer Research | 2007

Activation of Integrin-Linked Kinase Is a Critical Prosurvival Pathway Induced in Leukemic Cells by Bone Marrow–Derived Stromal Cells

Yoko Tabe; Linhua Jin; Yuko Tsutsumi-Ishii; Yuanyuan Xu; Teresa McQueen; Waldemar Priebe; Gordon B. Mills; Akimichi Ohsaka; Isao Nagaoka; Michael Andreeff; Marina Konopleva

Integrin-linked kinase (ILK) directly interacts with beta integrins and phosphorylates Akt in a phosphatidylinositol 3-kinase (PI3K)-dependent manner. In this study, we examined the functional role of ILK activation in leukemic and bone marrow stromal cells on their direct contact. Coculture of leukemic NB4 cells with bone marrow-derived stromal mesenchymal stem cells (MSC) resulted in robust activation of multiple signaling pathways, including ILK/Akt, extracellular signal-regulated kinase 1/2 (ERK1/2), signal transducers and activators of transcription 3 (STAT3), and Notch1/Hes. Blockade of PI3K or ILK signaling with pharmacologic inhibitors LY294002 or QLT0267 specifically inhibited stroma-induced phosphorylation of Akt and glycogen synthase kinase 3beta, suppressed STAT3 and ERK1/2 activation, and decreased Notch1 and Hes1 expression in leukemic cells. This resulted in induction of apoptosis in both leukemic cell lines and in primary acute myelogenous leukemia samples that was not abrogated by MSC coculture. In turn, leukemic cells growing in direct contact with bone marrow stromal elements induce activation of Akt, ERK1/2, and STAT3 signaling in MSC, accompanied by significant increase in Hes1 and Bcl-2 proteins, which were all suppressed by QLT0267 and LY294002. In summary, our results indicate reciprocal activation of ILK/Akt in both leukemic and bone marrow stromal cells. We propose that ILK/Akt is a proximal signaling pathway critical for survival of leukemic cells within the bone marrow microenvironment. Hence, disruption of these interactions by ILK inhibitors represents a potential novel therapeutic strategy to eradicate leukemia in the bone marrow microenvironment by simultaneous targeting of both leukemic cells and activated bone marrow stromal cells.


Clinical Cancer Research | 2009

MDM2 Antagonist Nutlin-3 Displays Antiproliferative and Proapoptotic Activity in Mantle Cell Lymphoma

Yoko Tabe; Denise Sebasigari; Linhua Jin; Martina Rudelius; Theresa Davies-Hill; Kazunori Miyake; Takashi Miida; Stefania Pittaluga; Mark Raffeld

Purpose: Mantle cell lymphoma (MCL) has one of the poorest prognoses of the non-Hodgkins lymphomas, and novel therapeutic approaches are needed. We wished to determine whether Nutlin-3, a novel small-molecule murine double minute 2 (MDM2) antagonist that efficiently activates TP53, might be effective in inducing cell death in MCL. Experimental Design: MCL cell lines with known TP53 status were treated with Nutlin-3, and biological and biochemical consequences were studied. Synergies with the prototypic genotoxic agent doxorubicin and the novel proteasome inhibitor bortezomib were assessed. Results: Nutlin-3 resulted in a reduction in cell proliferation/viability (IC50 < 10 μmol/L), an increase in the apoptotic fraction, and cell cycle arrest in wild-type (wt) TP53 Z-138 and Granta 519 cells. These effects were accompanied by TP53 accumulation and induction of TP53-dependent proteins p21, MDM2, Puma, and Noxa. Cell cycle arrest was characterized by suppression of S phase and an increase in the G0-G1 and G2-M fractions and accompanied by suppression of total and phosphorylated retinoblastoma protein and a decrease in G2-M-associated proteins cyclin B and CDC2. The combination of Nutlin-3 with doxorubicin or bortezomib was synergistic in wt-TP53 MCL cells. Nutlin-3 also induced cell cycle arrest and reduced cell viability in the mutant TP53 MINO cells but at a significantly higher IC50 (22.5 μmol/L). These effects were associated with induction of the TP53 homologue p73, slight increases in p21 and Noxa, and caspase activation. Nutlin-3 and bortezomib synergistically inhibited cell growth of MINO. Conclusion: These findings suggest that the MDM2 antagonist Nutlin-3 may be an effective agent in the treatment of MCL with or without wt-TP53.


Cell Death & Differentiation | 2007

Novel role of HDAC inhibitors in AML1/ETO AML cells: activation of apoptosis and phagocytosis through induction of annexin A1

Yoko Tabe; Linhua Jin; Rooha Contractor; David Gold; Peter P. Ruvolo; Susanne Radke; Yuanyuan Xu; Y. Tsutusmi-Ishii; Kazunori Miyake; Noriko Miyake; Sigemi Kondo; Akimichi Ohsaka; Isao Nagaoka; Michael Andreeff; Marina Konopleva

The chimeric fusion protein AML1-ETO, created by the t(8;21) translocation, recruits histone deacetylase (HDAC) to AML1-dependent promoters, resulting in transcriptional repression of the target genes. We analyzed the transcriptional changes in t(8;21) Kasumi-1 AML cells in response to the HDAC inhibitors, depsipeptide (FK228) and suberoylanilide hydroxamic acid (SAHA), which induced marked growth inhibition and apoptosis. Using cDNA array, annexin A1 (ANXA1) was identified as one of the FK228-induced genes. Induction of ANXA1 mRNA was associated with histone acetylation in ANXA1 promoter and reversal of the HDAC-dependent suppression of C/EBPα by AML1-ETO with direct recruitment of C/EBPα to ANXA1 promoter. This led to increase in the N-terminal cleaved isoform of ANXA1 protein and accumulation of ANXA1 on cell membrane. Neutralization with anti-ANXA1 antibody or gene silencing with ANXA1 siRNA inhibited FK228-induced apoptosis, suggesting that the upregulation of endogenous ANXA1 promotes cell death. FK228-induced ANXA1 expression was associated with massive increase in cell attachment and engulfment of Kasumi-1 cells by human THP-1-derived macrophages, which was completely abrogated with ANXA1 knockdown via siRNA transfection or ANXA1 neutralization. These findings identify a novel mechanism of action of HDAC inhibitors, which induce the expression and externalization of ANXA1 in leukemic cells, which in turn mediates the phagocytic clearance of apoptotic cells by macrophages.


Leukemia | 2012

Role of stromal microenvironment in nonpharmacological resistance of CML to imatinib through Lyn/CXCR4 interactions in lipid rafts

Yoko Tabe; Linhua Jin; Kazuhisa Iwabuchi; Rui-Yu Wang; Naoki Ichikawa; Takashi Miida; Jorge Cortes; Michael Andreeff; Marina Konopleva

We and others have previously demonstrated that p210 Bcr-Abl tyrosine kinase inhibits stromal cell-derived factor-1α/CXCR4 chemokine receptor signaling, contributing to the deficient adhesion of chronic myeloid leukemia (CML) cells to bone marrow stroma. Conversely, exposure of CML cells to a tyrosine kinase inhibitor (TKI) enhances migration of CML cells towards stromal cell layers and promotes non-pharmacological resistance to imatinib. Src-related kinase Lyn is known to interact with CXCL12/CXCR4 signaling and is directly activated by p210 Bcr-Abl. In this study, we demonstrate that TKI treatment promoted CXCR4 redistribution into the lipid raft fraction, in which it co-localized with active phosphorylated form of Lyn (LynTyr396) in CML cells. Lyn inhibition or cholesterol depletion abrogated imatinib-induced migration, and dual Src/Abl kinase inhibitor dasatinib induced fewer CML cells to migrate to the stroma. These findings demonstrate the novel mechanism of microenvironment-mediated resistance through lipid raft modulation, which involves compartmental changes of the multivalent CXCR4 and Lyn complex. We propose that pharmacological targeting of lipid rafts may eliminate bone marrow-resident CML cells through interference with microenvironment-mediated resistance.


PLOS ONE | 2013

TGF-β-Neutralizing Antibody 1D11 Enhances Cytarabine-Induced Apoptosis in AML Cells in the Bone Marrow Microenvironment.

Yoko Tabe; Yue Xi Shi; Zhihong Zeng; Linhua Jin; Masato Shikami; Yasuhito Hatanaka; Takashi Miida; Frank J. Hsu; Michael Andreeff; Marina Konopleva

Hypoxia and interactions with bone marrow (BM) stromal cells have emerged as essential components of the leukemic BM microenvironment in promoting leukemia cell survival and chemoresistance. High levels of transforming growth factor beta 1 (TGFβ1) produced by BM stromal cells in the BM niche regulate cell proliferation, survival, and apoptosis, depending on the cellular context. Exogenous TGFβ1 induced accumulation of acute myeloid leukemia (AML) cells in a quiescent G0 state, which was further facilitated by the co-culture with BM-derived mesenchymal stem cells (MSCs). In turn, TGFβ-neutralizing antibody 1D11 abrogated rhTGFβ1 induced cell cycle arrest. Blocking TGFβ with 1D11 further enhanced cytarabine (Ara-C)–induced apoptosis of AML cells in hypoxic and in normoxic conditions. Additional constituents of BM niche, the stroma-secreted chemokine CXCL12 and its receptor CXCR4 play crucial roles in cell migration and stroma/leukemia cell interactions. Treatment with 1D11 combined with CXCR4 antagonist plerixafor and Ara-C decreased leukemia burden and prolonged survival in an in vivo leukemia model. These results indicate that blockade of TGFβ by 1D11 and abrogation of CXCL12/CXCR4 signaling may enhance the efficacy of chemotherapy against AML cells in the hypoxic BM microenvironment.


Leukemia | 2010

Selective FLT3 inhibitor FI-700 neutralizes Mcl-1 and enhances p53-mediated apoptosis in AML cells with activating mutations of FLT3 through Mcl-1/Noxa axis.

Kensuke Kojima; Marina Konopleva; Twee Tsao; Michael Andreeff; H. Ishida; Y. Shiotsu; Linhua Jin; Yoko Tabe; Hideki Nakakuma

Treatment using Fms-like tyrosine kinase-3 (FLT3) inhibitors is a promising approach to overcome the dismal prognosis of acute myeloid leukemia (AML) with activating FLT3 mutations. Current trials are combining FLT3 inhibitors with p53-activating conventional chemotherapy. The mechanisms of cytotoxicity of FLT3 inhibitors are poorly understood. We investigated the interaction of FLT3 and p53 pathways after their simultaneous blockade using the selective FLT3 inhibitor FI-700 and the MDM2 inhibitor Nutlin-3 in AML. We found that FI-700 immediately reduced antiapoptotic Mcl-1 levels and enhanced Nutlin-induced p53-mediated mitochondrial apoptosis in FLT3/internal tandem duplication cells through the Mcl-1/Noxa axis. FI-700 induced proteasome-mediated degradation of Mcl-1, resulting in the reduced ability of Mcl-1 to sequester proapoptotic Bim. Nutlin-3 induced Noxa, which displaced Bim from Mcl-1. The FI-700/Nutlin-3 combination profoundly activated Bax and induced apoptosis. Our findings suggest that FI-700 actively enhances p53 signaling toward mitochondrial apoptosis and that a combination strategy aimed at inhibiting FLT3 and activating p53 signaling could potentially be effective in AML.


British Journal of Haematology | 2008

TGF‐β receptor kinase inhibitor LY2109761 reverses the anti‐apoptotic effects of TGF‐β1 in myelo‐monocytic leukaemic cells co‐cultured with stromal cells

Yuanyuan Xu; Yoko Tabe; Linhua Jin; Julie C. Watt; Teresa McQueen; Akimichi Ohsaka; Michael Andreeff; Marina Konopleva

Transforming growth factor β1 (TGF‐β1) is an essential regulator of cell proliferation, survival and apoptosis, depending on the cellular context. TGF‐β1 is also known to affect cell‐to‐cell interactions between tumour cells and stromal cells. We investigated the role of TGF‐β1 in the survival of myelo‐monocytic leukaemia cell lines co‐cultured with bone marrow (BM)‐derived mesenchymal stem cells (MSC). Treatment with recombinant human (rh)TGF‐β1 inhibited spontaneous and cytarabine‐induced apoptosis in U937 cells, most prominently in U937 cells directly attached to MSCs. Conversely, the pro‐survival effects of TGF‐β1 were inhibited by LY2109761 or TGF‐β1 neutralizing antibody. rhTGF‐β1 increased pro‐survival phosphorylation of Akt, which was inhibited by LY2109761. The combination of rhTGF‐β1 and MSC co‐culture induced significant upregulation of C/EBPβ gene (CEBPB) and protein expression along with increased C/EBPβ liver‐enriched activating protein: liver‐enriched inhibitory protein ratio, suggesting the novel role of C/EBPβ in TGF‐β1‐mediated U937 cell survival in the context of stromal cell support. In summary, these results indicate that TGF‐β1 produced by BM stromal cells promotes the survival and chemoresistance of leukaemia cells under the direct cell‐to‐cell interactions. The blockade of TGF‐β signalling by LY2109761, which effectively inhibited the pro‐survival signalling, may enhance the efficacy of chemotherapy against myelo‐monocytic leukaemic cells in the BM microenvironment.


Cancer Letters | 2010

MDM2 antagonist Nutlin-3 enhances bortezomib-mediated mitochondrial apoptosis in TP53-mutated mantle cell lymphoma

Linhua Jin; Yoko Tabe; Kensuke Kojima; Yixin Zhou; Stefania Pittaluga; Marina Konopleva; Takashi Miida; Mark Raffeld

This study demonstrated a pronounced synergistic growth-inhibitory effect of an MDM2 inhibitor Nutlin-3 and a proteasome inhibitor bortezomib in mantle cell lymphoma (MCL) cells regardless of TP53 mutant status and innate bortezomib sensitivity. In the mutant TP53 MCL cells which are intrinsically resistant to bortezomib, the combination of Nutlin-3/bortezomib synergistically induced cytotoxicity through the mitochondrial apoptotic pathway mediated by transcription-independent upregulation of NOXA, sequestration of MCL-1, activation of BAX, BAK, caspase-9 and -3. In the bortezomib sensitive wild-type TP53 MCL cells, the Nutlin-3/bortezomib combination caused G0/G1 cell cycle arrest followed by the increase in apoptosis induction. These findings indicate potential therapeutic efficacy of Nutlin-3/bortezomib combination for the treatment of chemorefractory MCL.


Annals of Hematology | 2006

PML-RARα and AML1–ETO translocations are rarely associated with methylation of the RARβ2 promoter

Yoko Tabe; Marina Konopleva; Yutaka Kondo; Rooha Contractor; Linhua Jin; Vivian Ruvolo; Yuko Tsutsumi-Ishii; Kazunori Miyake; Noriko Miyake; Akimichi Ohsaka; Isao Nagaoka; Jean-Pierre Issa; Michael Andreeff

The acute promyelocytic leukemia-specific PML–RARα fusion protein is a dominant-negative transcriptional repressor of retinoic acid receptor (RAR) target genes, which recruits HDAC and corepressor proteins and inhibits coactivators. Another oncogenic transcription factor, AML1–ETO, was proposed to cause an HDAC-dependent repression of RAR target genes. The RAR target RARβ2 gene has been reported to be frequently silenced by hypermethylation in many types of cancer cells. We examined the methylation status of the RARβ2 and asked if demethylation could reverse ATRA resistance in ATRA-resistant PML–RARα and AML1–ETO-positive cells. PML–RARα positive NB4 and its ATRA-resistant subvariant MR2 and AML1–ETO expressing Kasumi-1 cells had heterozygous methylation of RARβ2. Although DNA methyltransferase inhibitor 5-aza-2′-deoxycytidine partially reversed RARβ2 CpG methylation in these cells, it did not significantly enhance ATRA-induced RARβ2 mRNA expression and induction of maturation. However, the histone acetylase inhibitor SAHA combined with ATRA significantly reactivated RARβ2 mRNA both in NB4 and MR2 cells with degradation of PML–RARα, which was associated with maturation. In contrast, SAHA did not affect AML1–ETO levels and failed to induce RARβ2 expression and maturation in Kasumi-1 cells. In primary AML samples, RARβ2 expression was uniformly low; however, no specific correlation was observed between the methylation of the RARβ2 gene and expression of the fusion proteins, PML–RARα, and AML1–ETO. These results demonstrate that oncogenic PML–RARα and AML1–ETO translocations are rarely associated with RARβ2 promoter methylation in primary AML samples.

Collaboration


Dive into the Linhua Jin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marina Konopleva

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael Andreeff

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kensuke Kojima

Wakayama Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge