Network


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

Hotspot


Dive into the research topics where Zhi-Jian Liu is active.

Publication


Featured researches published by Zhi-Jian Liu.


The Journal of Steroid Biochemistry and Molecular Biology | 2004

Concentration-dependent mitogenic and antiproliferative actions of 2-methoxyestradiol in estrogen receptor-positive human breast cancer cells.

Zhi-Jian Liu; Bao Ting Zhu

We compared in this study the effects of 2-methoxyestradiol (2-MeO-E(2)) on the growth of two estrogen receptor (ER)-negative human breast cancer cell lines (MDA-MB-231 and MDA-MB-435s) and two ER-positive human breast cancer cell lines (MCF-7 and T-47D). 2-MeO-E(2) exerted a concentration-dependent antiproliferative action in the ER-negative MDA-MB-231 and MDA-MB-435s cells. The presence or absence of exogenous 17beta-estradiol (E(2)) in the culture medium did not affect the potency and efficacy of 2-MeO-E(2)s antiproliferative action in these ER-negative cells. When the ER-positive MCF-7 and T-47D cells were cultured in a medium supplemented with 10nM of exogenous E(2), 2-MeO-E(2) at 750 nM to 2 microM concentrations exerted a similar antiproliferative effect. However, when the ER-positive cell lines were cultured in the absence of exogenous E(2), 2-MeO-E(2) at relatively low concentrations (10-750 nM) had a moderate mitogenic effect, with its apparent efficacy 75-80% of that of E(2). This mitogenic effect of 2-MeO-E(2) was ER-mediated and largely attributable to 2-MeO-E(2)s residual estrogenic activity on the basis of our following findings: (i) its effect was only manifested in the ER-positive cells but not in the ER-negative cells; (ii) its effect in the ER-positive cells was partially or fully abolished when exogenous E(2) was concomitantly present in the culture medium; (iii) 2-MeO-E(2) retained 1-2% of E(2)s binding affinity for the human ERalpha and ERbeta, and its mitogenic effect was inhibited in a concentration-dependent manner by ICI-182,780, a pure ER antagonist; and (iv) its effect was not due to its metabolic conversion to 2-hydroxyestradiol. Our timely findings are of importance to the on-going clinical trials designed to evaluate 2-MeO-E(2)s effectiveness for the treatment of different types (ER-positive or ER-negative) of human breast cancer. This knowledge will improve the design of clinical trials as well as the interpretation of clinical outcomes when 2-MeO-E(2) is used as a single agent therapy or as part of a combination therapy for human breast cancer.


Blood | 2011

Developmental differences in megakaryocytopoiesis are associated with up-regulated TPO signaling through mTOR and elevated GATA-1 levels in neonatal megakaryocytes

Zhi-Jian Liu; Joseph E. Italiano; Francisca Ferrer-Marín; Ravi Kumar Gutti; Matthew Bailey; Brandon Poterjoy; Lisa M. Rimsza; Martha Sola-Visner

Multiple observations support the existence of developmental differences in megakaryocytopoiesis. We have previously shown that neonatal megakaryocyte (MK) progenitors are hyperproliferative and give rise to MKs smaller and of lower ploidy than adult MKs. Based on these characteristics, neonatal MKs have been considered immature. The molecular mechanisms underlying these differences are unclear, but contribute to the pathogenesis of disorders of neonatal megakaryocytopoiesis. In the present study, we demonstrate that low-ploidy neonatal MKs, contrary to traditional belief, are more mature than adult low-ploidy MKs. These mature MKs are generated at a 10-fold higher rate than adult MKs, and result from a developmental uncoupling of proliferation, polyploidization, and terminal differentiation. This pattern is associated with up-regulated thrombopoietin (TPO) signaling through mammalian target of rapamycin (mTOR) and elevated levels of full-length GATA-1 and its targets. Blocking of mTOR with rapamycin suppressed the maturation of neonatal MKs without affecting ploidy, in contrast to the synchronous inhibition of polyploidization and cytoplasmic maturation in adult MKs. We propose that these mechanisms allow fetuses/neonates to populate their rapidly expanding bone marrow and intravascular spaces while maintaining normal platelet counts, but also set the stage for disorders restricted to fetal/neonatal MK progenitors, including the Down syndrome-transient myeloproliferative disorder and the thrombocytopenia absent radius syndrome.


International Journal of Experimental Pathology | 2006

Developmental abnormalities in multiple proliferative tissues of ApcMin/+ mice

Shaojin You; Masami Ohmori; Maria Marjorette O. Peña; Basel Nassri; Jovelyn Quiton; Ziad A Al-Assad; Lucy Liu; Patricia A. Wood; Sondra H. Berger; Zhi-Jian Liu; Michael D. Wyatt; Robert L. Price; Franklin G. Berger; William J. M. Hrushesky

Germ‐line mutation of the Apc gene has been linked to familial adenomatous polyposis (FAP) that predisposes to colon cancer. ApcMin/+ mice, heterozygous for the Apc gene mutation, progressively develop small intestinal tumours in a manner that is analogous to that observed in the colon of patients with FAP ( Su et al. 1992; Fodde et al. 1994; Moser et al. 1995 ). We have studied the effects of Apc gene mutation on murine intestinal and extra‐intestinal, proliferatively active tissues. We have contrasted the histology to that of the age‐ and sex‐matched wild‐type C57BL/6 mice. Histological assessment of the normal appearing intestinal mucosa demonstrates minimal change in size of crypts. In contrast, villi are longer in the ileum of ApcMin/+ mice relative to C57BL/6 mice at 12 and 15 weeks of age. Vigorous splenic haematopoiesis in ApcMin/+ mice was seen at 12 and 15 weeks of age, as reflected by marked splenomegaly, increased splenic haematopoietic cells and megakaryocytes. Peripheral blood counts, however, did not differ between C57BL/6 and ApcMin/+ mice at 15 weeks of age. Lymphoid depletion in ApcMin/+ mice was characterized by diminished numbers of splenic lymphoid follicles and small intestinal Peyers patches. The ovaries of 12‐ and 15‐week‐old ApcMin/+ mice exhibited increased numbers of atretic follicles, and estrous cycling by serial vaginal smears showed tendency of elongation in the mutant mice during these age ranges. The testicles of 10‐week‐old ApcMin/+ mice showed increased numbers of underdeveloped seminiferous tubules. Collectively, these data suggest that, in addition to its obvious effects upon intestinal adenoma formation, Apc gene mutation causes impairment of developmental and apparent differentiation blockade in proliferative tissues, including those of the haematopoietic system, lymphoid and reproductive tract.


Blood | 2014

Expansion of the neonatal platelet mass is achieved via an extension of platelet lifespan.

Zhi-Jian Liu; Karin M. Hoffmeister; Zhongbo Hu; Donald E. Mager; Sihem Ait-Oudhia; Marlyse A. Debrincat; Irina Pleines; Emma C. Josefsson; Benjamin T. Kile; Joseph E. Italiano; Haley Ramsey; Renata Grozovsky; Peter Veng-Pedersen; Chaitanya Chavda; Martha Sola-Visner

The fetal/neonatal hematopoietic system must generate enough blood cells to meet the demands of rapid growth. This unique challenge might underlie the high incidence of thrombocytopenia among preterm neonates. In this study, neonatal platelet production and turnover were investigated in newborn mice. Based on a combination of blood volume expansion and increasing platelet counts, the platelet mass increased sevenfold during the first 2 weeks of murine life, a time during which thrombopoiesis shifted from liver to bone marrow. Studies applying in vivo biotinylation and mathematical modeling showed that newborn and adult mice had similar platelet production rates, but neonatal platelets survived 1 day longer in circulation. This prolonged lifespan fully accounted for the rise in platelet counts observed during the second week of murine postnatal life. A study of pro-apoptotic and anti-apoptotic Bcl-2 family proteins showed that neonatal platelets had higher levels of the anti-apoptotic protein Bcl-2 and were more resistant to apoptosis induced by the Bcl-2/Bcl-xL inhibitor ABT-737 than adult platelets. However, genetic ablation or pharmacologic inhibition of Bcl-2 alone did not shorten neonatal platelet survival or reduce platelet counts in newborn mice, indicating the existence of redundant or alternative mechanisms mediating the prolonged lifespan of neonatal platelets.


Journal of Clinical Investigation | 2014

Proteasome function is required for platelet production

Dallas S. Shi; Matthew C. Smith; Robert A. Campbell; Patrick W. Zimmerman; Zechariah B. Franks; Bjorn F. Kraemer; Kellie R. Machlus; Jing Ling; Patrick Kamba; Hansjörg Schwertz; Jesse W. Rowley; Rodney R. Miles; Zhi-Jian Liu; Martha Sola-Visner; Joseph E. Italiano; Hilary Christensen; Walter H. A. Kahr; Dean Y. Li; Andrew S. Weyrich

The proteasome inhibiter bortezomib has been successfully used to treat patients with relapsed multiple myeloma; however, many of these patients become thrombocytopenic, and it is not clear how the proteasome influences platelet production. Here we determined that pharmacologic inhibition of proteasome activity blocks proplatelet formation in human and mouse megakaryocytes. We also found that megakaryocytes isolated from mice deficient for PSMC1, an essential subunit of the 26S proteasome, fail to produce proplatelets. Consistent with decreased proplatelet formation, mice lacking PSMC1 in platelets (Psmc1(fl/fl) Pf4-Cre mice) exhibited severe thrombocytopenia and died shortly after birth. The failure to produce proplatelets in proteasome-inhibited megakaryocytes was due to upregulation and hyperactivation of the small GTPase, RhoA, rather than NF-κB, as has been previously suggested. Inhibition of RhoA or its downstream target, Rho-associated protein kinase (ROCK), restored megakaryocyte proplatelet formation in the setting of proteasome inhibition in vitro. Similarly, fasudil, a ROCK inhibitor used clinically to treat cerebral vasospasm, restored platelet counts in adult mice that were made thrombocytopenic by tamoxifen-induced suppression of proteasome activity in megakaryocytes and platelets (Psmc1(fl/fl) Pdgf-Cre-ER mice). These results indicate that proteasome function is critical for thrombopoiesis, and suggest inhibition of RhoA signaling as a potential strategy to treat thrombocytopenia in bortezomib-treated multiple myeloma patients.


Nature Genetics | 2016

A missense mutation in TFRC, encoding transferrin receptor 1, causes combined immunodeficiency

Haifa H. Jabara; Steven E. Boyden; Janet Chou; Narayanaswamy Ramesh; Michel J. Massaad; Halli Benson; Wayne Bainter; David Fraulino; Fedik Rahimov; Colin A. Sieff; Zhi-Jian Liu; Salem H. Alshemmari; Basel K. al-Ramadi; Hasan Al-Dhekri; Rand Arnaout; Mohammad Abu-Shukair; Anant Vatsayan; Eli Silver; Sanjay Ahuja; E. Graham Davies; Martha Sola-Visner; Toshiro K. Ohsumi; Nancy C. Andrews; Luigi D. Notarangelo; Mark D. Fleming; Waleed Al-Herz; Louis M. Kunkel; Raif S. Geha

Patients with a combined immunodeficiency characterized by normal numbers but impaired function of T and B cells had a homozygous p.Tyr20His substitution in transferrin receptor 1 (TfR1), encoded by TFRC. The substitution disrupts the TfR1 internalization motif, resulting in defective receptor endocytosis and markedly increased TfR1 expression on the cell surface. Iron citrate rescued the lymphocyte defects, and expression of wild-type but not mutant TfR1 rescued impaired transferrin uptake in patient-derived fibroblasts. TfrcY20H/Y20H mice recapitulated the immunological defects of patients. Despite the critical role of TfR1 in erythrocyte development and function, patients had only mild anemia and only slightly increased TfR1 expression in erythroid precursors. We show that STEAP3, a metalloreductase expressed in erythroblasts, associates with TfR1 and partially rescues transferrin uptake in patient-derived fibroblasts, suggesting that STEAP3 may provide an accessory TfR1 endocytosis signal that spares patients from severe anemia. These findings demonstrate the importance of TfR1 in adaptive immunity.


Seminars in Hematology | 2010

Neonatal Thrombocytopenia and Megakaryocytopoiesis

Francisca Ferrer-Marín; Zhi-Jian Liu; Ravi Kumar Gutti; Martha Sola-Visner

Thrombocytopenia is common among sick neonates, affecting 20% to 35% of all patients admitted to the neonatal intensive care unit (NICU). While most cases of neonatal thrombocytopenia are mild or moderate and resolve within 7 to 14 days with appropriate therapy, 2.5% to 5% of NICU patients develop severe thrombocytopenia, sometimes lasting for several weeks and requiring >20 platelet transfusions. The availability of thrombopoietic agents offers the possibility of decreasing the number of platelet transfusions and potentially improving the outcomes of these infants. However, adding thrombopoietin (TPO) mimetics to the therapeutic armamentarium of neonatologists will require careful attention to the substantial developmental differences between neonates and adults in the process of megakaryocytopoiesis and in their responses to TPO. Taken together, the available data suggest that TPO mimetics will stimulate platelet production in neonates, but might do so through different mechanisms and at different doses than those established for adults. In addition, the specific groups of thrombocytopenic neonates most likely to benefit from therapy with TPO mimetics need to be defined, and the potential nonhematological effects of these agents on the developing organism need to be considered. This review summarizes our current understanding of neonatal megakaryocytopoiesis, and examines in detail the developmental factors relevant to the potential use of TPO mimetics in neonates.


Current Opinion in Hematology | 2011

Neonatal and adult megakaryopoiesis.

Zhi-Jian Liu; Martha Sola-Visner

Purpose of reviewIt has become increasingly clear that there are substantial biological differences between fetal/neonatal and adult megakaryopoiesis. Over the last 18 months, studies challenged the paradigm that neonatal megakaryocytes are immature and revealed a developmentally unique uncoupling of proliferation, polyploidization, and cytoplasmic maturation. Several studies also described substantial molecular differences between fetal/neonatal and adult megakaryocytes involving transcription factors, signaling pathways, cytokine receptors, and microRNAs. Recent findingsThis review will summarize our current knowledge on the developmental differences between fetal/neonatal and adult megakaryocytes, and recent advances in the underlying molecular mechanisms, including differences in transcription factors, in the response to thrombopoietin (Tpo), and newly described developmentally regulated signaling pathways. We will also discuss the implications of these findings on the way megakaryocytes interact with the environment, the response of neonates to thrombocytopenia, and the pathogenesis of Down syndrome-transient myeloproliferative disorder (TMD) and Down syndrome-acute megakaryoblastic leukemia (DS-AMKL). SummaryA better characterization of the molecular differences between fetal/neonatal and adult megakaryocytes is critical to elucidating the pathogenesis of a group of disorders that selectively affect fetal/neonatal megakaryocyte progenitors, including the thrombocytopenia-absent radius (TAR) syndrome, Down syndrome-TMD or Down syndrome-AMKL, and the delayed platelet engraftment following cord blood transplantation.


Journal of Perinatology | 2010

Increasing platelets without transfusion: is it time to introduce novel thrombopoietic agents in neonatal care?

Hannes Sallmon; Ravi Kumar Gutti; Francisca Ferrer-Marín; Zhi-Jian Liu; Martha Sola-Visner

The Food and Drug Administration recently approved two novel thrombopoiesis-stimulating agents, Romiplostim (AMG-531, Nplate) and Eltrombopag (Promacta), for the treatment of adults with immune thrombocytopenic purpura. For physicians taking care of critically ill neonates, this offers the opportunity of decreasing platelet transfusions and potentially improving the outcomes of neonates with severe and prolonged thrombocytopenia. However, several developmental factors need to be taken into consideration. First, the population of thrombocytopenic neonates likely to benefit from these agents needs to be carefully selected. Second, the mechanisms underlying neonatal and adult thrombocytopenia differ from each other and are incompletely understood, and pre-clinical evidence suggests that the response of neonates to thrombopoietic factors might be different from that of adults. Finally, the potential non-hematopoietic effects of thrombopoietin have not been well established. Here, we will discuss these issues in detail, and will highlight the critical developmental differences between neonates and adults that need to be considered as we think about introducing these agents into neonatal care.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2012

The hibernating 13-lined ground squirrel as a model organism for potential cold storage of platelets

Scott T. Cooper; Karl E. Richters; Travis E. Melin; Zhi-Jian Liu; Peter J. Hordyk; Ryan R. Benrud; Lauren R. Geiser; Steve E. Cash; C. Simon Shelley; David R. Howard; Mark H. Ereth; Martha Sola-Visner

Hibernating mammals have developed many physiological adaptations to extreme environments. During hibernation, 13-lined ground squirrels (Ictidomys tridecemlineatus) must suppress hemostasis to survive prolonged body temperatures of 4-8°C and 3-5 heartbeats per minute without forming lethal clots. Upon arousal in the spring, these ground squirrels must be able to quickly restore normal clotting activity to avoid bleeding. Here we show that ground squirrel platelets stored in vivo at 4-8°C were released back into the blood within 2 h of arousal in the spring with a body temperature of 37°C but were not rapidly cleared from circulation. These released platelets were capable of forming stable clots and remained in circulation for at least 2 days before newly synthesized platelets were detected. Transfusion of autologous platelets stored at 4°C or 37°C showed the same clearance rates in ground squirrels, whereas rat platelets stored in the cold had a 140-fold increase in clearance rate. Our results demonstrate that ground squirrel platelets appear to be resistant to the platelet cold storage lesions observed in other mammals, allowing prolonged storage in cold stasis and preventing rapid clearance upon spring arousal. Elucidating these adaptations could lead to the development of methods to store human platelets in the cold, extending their shelf life.

Collaboration


Dive into the Zhi-Jian Liu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph E. Italiano

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Chaitanya Chavda

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Haley Ramsey

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karin M. Hoffmeister

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Viola Lorenz

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge