Network


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

Hotspot


Dive into the research topics where Alexander L. Kovalchuk is active.

Publication


Featured researches published by Alexander L. Kovalchuk.


Proceedings of the National Academy of Sciences of the United States of America | 2002

IL-6 transgenic mouse model for extraosseous plasmacytoma

Alexander L. Kovalchuk; Joong Su Kim; Sung Sup Park; Allen E. Coleman; Jerrold M. Ward; Herbert C. Morse; Tadamitsu Kishimoto; Michael Potter; Siegfried Janz

Plasma cell neoplasms in humans comprise plasma cell myeloma, otherwise known as multiple myeloma, Ig deposition and heavy chain diseases, and plasmacytoma (PCT). A subset of PCT, designated extramedullary PCT, is distinguished from multiple myeloma and solitary PCT of bone by its distribution among various tissue sites but not the bone marrow. Extramedullary (extraosseus) PCT are rare spontaneous neoplasms of mice but are readily induced in a susceptible strain, BALB/c, by treatment with pristane. The tumors develop in peritoneal granulomas and are characterized by Myc-activating T(12;15) chromosomal translocations and, most frequently, by secretion of IgA. A uniting feature of human and mouse plasma cell neoplasms is the critical role played by IL-6, a B cell growth, differentiation, and survival factor. To directly test the contribution of IL-6 to PCT development, we generated BALB/c mice carrying a widely expressed IL-6 transgene. All mice exhibited lymphoproliferation and plasmacytosis. By 18 months of age, over half developed readily transplantable PCT in lymph nodes, Peyers patches, and sometimes spleen. These neoplasms also had T(12;15) translocations, but remarkably, none expressed IgA. Unexpectedly, ≈30% of the mice developed follicular and diffuse large cell B cell lymphomas that often coexisted with PCT. These findings provide a unique model of extramedullary PCT for studies on pathogenesis and treatment and suggest a previously unappreciated role for IL-6 in the genesis of germinal center-derived lymphomas.


Journal of Experimental Medicine | 2007

AID-deficient Bcl-xL transgenic mice develop delayed atypical plasma cell tumors with unusual Ig/Myc chromosomal rearrangements

Alexander L. Kovalchuk; Wendy Dubois; Elizabeth Mushinski; Nicole McNeil; Carsten Hirt; Chen-Feng Qi; Zhaoyang Li; Siegfried Janz; Tasuku Honjo; Masamichi Muramatsu; Thomas Ried; Timothy W. Behrens; Michael Potter

Activation-induced cytidine deaminase (AID) is required for immunoglobulin (Ig) class switch recombination and somatic hypermutation, and has also been implicated in translocations between Ig switch regions and c-Myc in plasma cell tumors in mice. We asked if AID is required for accelerated tumor development in pristane-treated Bcl-xL transgenic BALB/c mice deficient in AID (pBxAicda−/−). pBxAicda −/− mice developed tumors with a lower frequency (24 vs. 62%) and a longer mean latency (108 vs. 36 d) than AID-sufficient mice. The tumors appeared in oil granuloma tissue and did not form ascites. By interphase fluorescence in situ hybridization, six out of nine pBxAicda −/− primary tumors had T(12;15) and one had T(6;15) chromosomal translocations. Two tumors were transplantable and established as stable cell lines. Molecular and cytogenetic analyses showed that one had an unusual unbalanced T(12;15) translocation, with IgH Cμ and Pvt-1 oriented head to tail at the breakpoint, resulting in an elevated expression of c-Myc. In contrast, the second was T(12;15) negative, but had an elevated N-Myc expression caused by a paracentric inversion of chromosome 12. Thus, novel mechanisms juxtapose Ig and Myc-family genes in AID-deficient plasma cell tumors.


Oncogene | 1997

Deletional remodeling of c-myc-deregulating chromosomal translocations

Alexander L. Kovalchuk; Jürgen Müller; Siegfried Janz

Evidence is presented for the existence of a novel remodeling-by-deletion mechanism that alters the fine structure of c-myc-deregulating chromosomal translocations in t(12;15)-positive BALB/c plasmacytomas. DNA sequence analysis of the t(12;15) in five primary tumors revealed the co-existence of precursor cells harboring genetic recombinations between the immunoglobulin heavy-chain μ locus (Ighμ) and c-myc with clonally related progenitors containing rearrangements between the immunoglobulin heavy-chain α locus (Ighα) and c-myc. Clonal relatedness was based upon unique junction fragments between the switch region of Ighμ and c-myc. Sμ/c-myc junctions are thus useful clonotypic markers for monitoring the conversion of Ighμ/c-myc-positive tumor precursor clones into Ighα/c-myc-positive plasmacytomas. Aberrant isotype switch recombination appears to be the most likely mechanism effecting this conversion event (other possibilities are discussed) which may help to explain the preferred usage of the Ighα locus in recombinations with c-myc in t(12;15)-positive plasma cell tumors in BALB/c mice.


Journal of Experimental Medicine | 2002

Disruption of transforming growth factor β signaling by a novel ligand-dependent mechanism

Tania Fernandez; Stephanie R. Amoroso; Shellyann Sharpe; Gary Jones; Valery Bliskovski; Alexander L. Kovalchuk; Lalage M. Wakefield; Seong-Jin Kim; Michael Potter; John J. Letterio

Transforming growth factor (TGF)-β is the prototype in a family of secreted proteins that act in autocrine and paracrine pathways to regulate cell development and function. Normal cells typically coexpress TGF-β receptors and one or more isoforms of TGF-β, thus the synthesis and secretion of TGF-β as an inactive latent complex is considered an essential step in regula-ting the activity of this pathway. To determine whether intracellular activation of TGF-β results in TGF-β ligand–receptor interactions within the cell, we studied pristane-induced plasma cell tumors (PCTs). We now demonstrate that active TGF-β1 in the PCT binds to intracellular TGF-β type II receptor (TβRII). Disruption of the expression of TGF-β1 by antisense TGF-β1 mRNA restores localization of TβRII at the PCT cell surface, indicating a ligand-induced impediment in receptor trafficking. We also show that retroviral expression of a truncated, dominant-negative TβRII (dnTβRII) effectively competes for intracellular binding of active ligand in the PCT and restores cell surface expression of the endogenous TβRII. Analysis of TGF-β receptor–activated Smad2 suggests the intracellular ligand–receptor complex is not capable of signaling. These data are the first to demonstrate the formation of an intracellular TGF-β–receptor complex, and define a novel mechanism for modulating the TGF-β signaling pathway.


PLOS ONE | 2010

Eef1a2 Promotes Cell Growth, Inhibits Apoptosis and Activates JAK/STAT and AKT Signaling in Mouse Plasmacytomas

Zhaoyang Li; Chen-Feng Qi; Dong-Mi Shin; Adriana Zingone; Helen J. Newbery; Alexander L. Kovalchuk; Catherine M. Abbott; Herbert C. Morse

Background The canonical function of EEF1A2, normally expressed only in muscle, brain, and heart, is in translational elongation, but recent studies suggest a non-canonical function as a proto-oncogene that is overexpressed in a variety of solid tumors including breast and ovary. Transcriptional profiling of a spectrum of primary mouse B cell lineage neoplasms showed that transcripts encoding EEF1A2 were uniquely overexpressed in plasmacytomas (PCT), tumors of mature plasma cells. Cases of human multiple myeloma expressed significantly higher levels of EEF1A2 transcripts than normal bone marrow plasma cells. High-level expression was also a feature of a subset of cell lines developed from mouse PCT and from the human MM. Methodology/Principal Findings Heightened expression of EEF1A2 was not associated with increased copy number or coding sequence mutations. shRNA-mediated knockdown of Eef1a2 transcripts and protein was associated with growth inhibition due to delayed G1-S progression, and effects on apoptosis that were seen only under serum-starved conditions. Transcriptional profiles and western blot analyses of knockdown cells revealed impaired JAK/STAT and PI3K/AKT signaling suggesting their contributions to EEF1A2-mediated effects on PCT induction or progression. Conclusions/Significance EEF1A2 may play contribute to the induction or progression of some PCT and a small percentage of MM. Eef1a2 could also prove to be a useful new marker for a subset of MM and, ultimately, a possible target for therapy.


Cell Cycle | 2013

Rap2b, a novel p53 target, regulates p53-mediated pro-survival function.

Xinyue Zhang; Yunlong He; Kyoung-Hwa Lee; Wendy Dubois; Ziqing Li; Xiaolin Wu; Alexander L. Kovalchuk; Weimin Zhang; Jing Huang

The tumor suppressor p53 is a critical regulator of apoptosis and cell cycle arrest/pro-survival. Upon DNA damage, p53 evokes both cell cycle arrest/pro-survival and apoptosis transcriptional programs. The ultimate cellular outcome depends on the balance of these two programs. However, the p53 downstream targets that mediate this cell fate decision remain to be identified. Using an integrative genomic approach, we identify Rap2b as a conserved p53-activated gene that counters p53-mediated apoptosis after DNA damage. Upon DNA damage, p53 directly binds to the promoter of Rap2b and activates its transcription. The reduction of Rap2b levels by small interference RNA sensitizes cells to DNA damage-induced apoptosis in a p53-dependent manner. Consistent with its pro-survival function, analysis of cancer genomic data reveals that Rap2b is overexpressed in many types of tumors. Anchorage-independent growth assays show that Rap2b has only weak transformation activity, suggesting that it is not an oncogene by itself. Together, our results identify Rap2b as a new player in the pro-survival program conducted by p53 and raise the possibility that targeting Rap2b could sensitize tumor cells to apoptosis in response to DNA damage.


Cancer Research | 2007

Anaplastic, plasmablastic, and plasmacytic plasmacytomas of mice: relationships to human plasma cell neoplasms and late-stage differentiation of normal B cells.

Chen-Feng Qi; Jeff X. Zhou; Chang Hoon Lee; Zohreh Naghashfar; Shao Xiang; Alexander L. Kovalchuk; Torgny N. Fredrickson; Janet W. Hartley; Derry C. Roopenian; Wendy F. Davidson; Siegfried Janz; Herbert C. Morse

We have compared histologic features and gene expression profiles of newly identified plasmacytomas from NFS.V(+) congenic mice with plasmacytomas of IL6 transgenic, Fasl mutant, and SJL-beta2M(-/-) mice. NFS.V(+) tumors comprised an overlapping morphologic spectrum of high-grade/anaplastic, intermediate-grade/plasmablastic, and low-grade/plasmacytic cases with similarities to subsets of human multiple myeloma and plasmacytoma. Microarray and immunohistochemical analyses of genes expressed by the most prevalent tumors, plasmablastic plasmacytomas, showed them to be most closely related to immunoblastic lymphomas, less so to plasmacytomas of Fasl mutant and SJL mice, and least to plasmacytic plasmacytomas of IL6 transgenic mice. Plasmablastic tumors seemed to develop in an inflammatory environment associated with gene signatures of T cells, natural killer cells, and macrophages not seen with plasmacytic plasmacytomas. Plasmablastic plasmacytomas from NFS.V(+) and SJL-beta2M(-/-) mice did not have structural alterations in Myc or T(12;15) translocations and did not express Myc at high levels, regular features of transgenic and pristane-induced plasmacytomas. These findings imply that, as for human multiple myeloma, Myc-independent routes of transformation contribute to the pathogenesis of these tumors. These findings suggest that plasma cell neoplasms of mice and humans exhibit similar degrees of complexity. Mouse plasmacytomas, previously considered to be homogeneous, may thus be as diverse as their human counterparts with respect to oncogenic mechanisms of plasma cell transformation. Selecting specific types of mouse plasmacytomas that relate most closely to subtypes of human multiple myeloma may provide new opportunities for preclinical testing of drugs for treatment of the human disease.


Oncogene | 2002

Genomic instability in mouse Burkitt lymphoma is dominated by illegitimate genetic recombinations, not point mutations.

Lynne D Rockwood; Ted A. Torrey; Joong Su Kim; Allen E. Coleman; Alexander L. Kovalchuk; Shao Xiang; Thomas Ried; Herbert C. Morse; Siegfried Janz

λ-MYC-induced mouse Burkitt lymphoma (BL) harboring the shuttle vector pUR288, which includes a lacZ reporter gene to study mutagenesis, was employed to assess genomic instability associated with MYC deregulation. The frequency of lacZ mutations in lymphomas was elevated only 1.75-fold above that in normal tissue, indicating that mouse BL does not exhibit a phenotype of hypermutability. However, the nature of lacZ mutations was strikingly different in normal tissues and lymphomas. While point mutations comprised approximately 75% of the mutations found in normal tissues, apparent translocations, deletions and inversions constituted the majority of mutations (∼65%) in lymphomas. Genomic instability in mouse BL thus seems characterized by a preponderance of illegitimate genetic rearrangements in the context of near-background mutant frequencies. SKY analyses of cell lines from primary BL tumors revealed substantial changes in chromosomal structure, confirming the lacZ studies. Bi-allelic deletions of the tumor suppressor p16Ink4a were detected in six out of 16 cell lines, illustrating cellular selection of advantageous mutations. Together, these approaches indicate that MYC may contribute to lymphomagenesis through the dominant mutator effect of inducing chromosomal instability. The results further suggest that a phenotype of hypermutability (elevated mutant frequency) may not always be required for oncogenesis to occur.


Leukemia | 2000

Lymph nodes and Peyer's patches of IL-6 transgenic BALB/c mice harbor T(12;15) translocated plasma cells that contain illegitimate exchanges between the immunoglobulin heavy-chain μ locus and c-myc

Alexander L. Kovalchuk; Tadamitsu Kishimoto; Siegfried Janz

Hyperplastic plasmacytotic lymph nodes and Peyers patches of 12 of 25 (48%) BALB/c mice that carried a human IL-6 transgene under the transcriptional control of the histocompatibility H-2LD promoter (BALB/c.IL-6 mice) were found to harbor 15 cell clones that contained in their T(12;15) translocation breakpoint regions illegitimate genetic recombinations between the upstream flank of the immunoglobulin heavy-chain Cμ locus (5′-Cμ) and c-myc(5′-Cμ/c-myc+ clones). Similar 5′-Cμ/c-myc+ clones were also detected in pristane-induced peritoneal granulomata (a significant source of IL-6 in situ) of three of 13 (13%) conventional BALB/c mice, but not in lymphoid tissues of pristane-treated BALB/c mice, nor in any tissue of untreated BALB/c mice. These findings provided strong evidence that IL-6 may be able to promote the growth and/or survival of clones that contained rearrangements between 5′-Cμ and c-myc. Taken in conjunction with our previous observation that 5′-Cμ/c-myc+ clones are the precursors for pristane-induced BALB/c plasmacytomas, the findings further suggested that IL-6 may play a pivotal role in the early stage of plasmacytoma development, by promoting tumor precursor cells. The BALB/c.IL-6 model of plasmacytomagenesis may be superior to the conventional BALA/c model because the putative plasmacytoma precursors appear to be more prevalent and in their development independent of treating the mice with inflammation-inducing plasmacytomagenic agents, such as pristane or silicone polymers.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Expression of plasma cell alloantigen 1 defines layered development of B-1a B-cell subsets with distinct innate-like functions.

Hongsheng Wang; Dong-Mi Shin; Sadia Abbasi; Shweta Jain; Alexander L. Kovalchuk; Natalie Beaty; Sophia Chen; Ines Gonzalez-Garcia; Herbert C. Morse

Innate-like B-1a cells contribute significantly to circulating natural antibodies and mucosal immunity as well as to immunoregulation. Here we show that these classic functions of B-1a cells segregate between two unique subsets defined by expression of plasma cell alloantigen 1 (PC1), also known as ectonucleotide pyrophosphatase phosphodiesterase 1 (ENPP1). These subsets, designated B-1a.PC1lo and B-1a.PC1hi, differ significantly in IgH chain utilization. Adoptively transferred PC1lo cells secreted significantly more circulating natural IgM and intestinal IgA than PC1hi cells. In contrast, PC1hi cells produced more IL-10 than PC1lo cells when stimulated with LPS and phorbol 12-myristate 13-acetate (PMA). PC1hi cells were also more efficient than PC1lo cells in regulating Th1 cell differentiation, even though both B-1a subsets were comparably active in stimulating T-cell proliferation. Furthermore, PC1lo cells generated antigen-specific IgM responses to pneumococcal polysaccharide antigens, whereas PC1hi cells do not. We found that PC1lo cells develop from an early wave of B-1a progenitors in fetal life, whereas PC1hi cells are generated from a later wave after birth. We conclude that identification of B-1a.PC1lo and B-1a.PC1hi cells extends the concept of a layered immune system with important implications for developing effective vaccines and promoting the generation of immunoregulatory B cells.

Collaboration


Dive into the Alexander L. Kovalchuk's collaboration.

Top Co-Authors

Avatar

Herbert C. Morse

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Siegfried Janz

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Chen-Feng Qi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Hongsheng Wang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Dong-Mi Shin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Wendy Dubois

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Zhaoyang Li

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Michael Potter

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Allen E. Coleman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Shao Xiang

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge