Network


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

Hotspot


Dive into the research topics where Jeffrey H. Albrecht is active.

Publication


Featured researches published by Jeffrey H. Albrecht.


Oncogene | 1998

Involvement of p21 and p27 in the regulation of CDK activity and cell cycle progression in the regenerating liver

Jeffrey H. Albrecht; Randy Yat Choi Poon; Cory L. Ahonen; Brenda M. Rieland; Chu-Xia Deng; Gretchen S. Crary

In tissue culture systems, p21 and p27 inhibit cyclin-dependent kinase (CDK) activity and cell cycle progression in response to numerous stimuli, but little is known about their involvement in cell growth in vivo. We examined the modulation of CDK activity by these proteins after 70% partial hepatectomy (PH), an in vivo model of synchronous hepatocyte cell cycle progression. After PH in BALB/c mice, p21 was induced during the prereplicative (G1) phase and was maximally expressed after peak hepatocyte DNA synthesis. p27 was present in quiescent liver and was minimally induced after PH. p21 and p27 immunoprecipitated with CDK2, CDK4, and cyclin D1 in the regenerating liver. The activity of CDK2-, CDK4- and cyclin D1-associated kinases was upregulated after PH, and maximal activity of these enzyme complexes corresponded to peak DNA synthesis. Immunodepletion experiments suggested that p27 plays a role in downregulating CDK2 activity before and after peak DNA synthesis. Compared to cogenic wild-type mice, p21−/− mice demonstrated evidence of markedly accelerated hepatocyte progression through G1 phase after PH: DNA synthesis, upregulation of cyclin A and PCNA, induction of cyclin D1- and CDK2-associated kinase activity, and appearance of a phosphorylated retinoblastoma protein (Rb) species occurred earlier in the p21−/− mice. These results suggest that p21 and p27 modulate CDK activity in the regenerating liver, and that p21 regulates the rate of progression through G1 phase of the cell cycle in vivo.


Journal of Clinical Oncology | 2006

Prospective Multi-Institutional Study of Reverse Transcriptase Polymerase Chain Reaction for Molecular Staging of Melanoma

Charles R. Scoggins; Merrick I. Ross; Douglas S. Reintgen; R. Dirk Noyes; James S. Goydos; Peter D. Beitsch; Marshall M. Urist; Stephan Ariyan; B. Scott Davidson; Jeffrey J. Sussman; Michael J. Edwards; Robert C.G. Martin; Angela M. Lewis; Arnold J. Stromberg; Andrew Conrad; Lee Hagendoorn; Jeffrey H. Albrecht; Kelly M. McMasters

PURPOSE To evaluate the prognostic significance of molecular staging using reverse transcriptase polymerase chain reaction (RT-PCR) in detecting occult melanoma cells in sentinel lymph nodes (SLNs) and circulating bloodstream. PATIENTS AND METHODS In this multicenter study, eligibility criteria included patient age 18 to 71 years, invasive melanoma > or = 1.0 mm Breslow thickness, and no clinical evidence of metastasis. SLN biopsy and wide excision of the primary tumor were performed. SLNs were examined by serial-section histopathology and S-100 immunohistochemistry. A portion of each SLN was frozen for RT-PCR. In addition, RT-PCR was performed on peripheral-blood mononuclear cells (PBMCs). RT-PCR analysis was performed using four markers: tyrosinase, MART1, MAGE3, and GP-100. Disease-free survival (DFS), distant-DFS (DDFS), and overall survival (OS) were analyzed. RESULTS A total of 1,446 patients with histologically negative SLNs underwent RT-PCR analysis. At a median follow-up of 30 months, there was no difference in DFS, DDFS, or OS between the RT-PCR-positive (n = 620) and RT-PCR-negative (n = 826) patients. Analysis of PBMC from 820 patients revealed significant differences in DFS and DDFS, but not OS, for patients with detection of more than one RT-PCR marker in peripheral blood. CONCLUSION In this large, prospective, multi-institutional study, RT-PCR analysis on SLNs and PBMCs provides no additional prognostic information beyond standard histopathologic analysis of SLNs. Detection of more than one marker in PBMC is associated with a worse prognosis. RT-PCR remains investigational and should not be used to direct adjuvant therapy at this time.


Journal of Biological Chemistry | 2003

Evidence That Cyclin D1 Mediates Both Growth and Proliferation Downstream of TOR in Hepatocytes

Christopher J. Nelsen; David G. Rickheim; Melissa M. Tucker; Linda K. Hansen; Jeffrey H. Albrecht

Signaling through the target of rapamycin is required for increased protein synthesis, cell growth, and proliferation in response to growth factors. However, the downstream mediators of these responses, and the elements linking growth and proliferation, have not been fully elucidated. Rapamycin inhibits hepatocyte proliferation in culture and liver regenerationin vivo. In cultured rat hepatocytes, rapamycin prevented the up-regulation of cyclin D1 as well as proteins acting downstream in the cell cycle. Transfection with cyclin D1 or E2F2, but not cyclin E or activated Akt, overcame the rapamycin-mediated cell cycle arrest. Rapamycin also inhibited the induction of global protein synthesis after growth factor stimulation, and cyclin D1 overcame this inhibition. Rapamycin inhibited hepatocyte proliferation and cyclin D1 expression in the mouse liver after 70% partial hepatectomy. In rapamycin-treated mice, transfection with cyclin D1 induced hepatocyte proliferation, increased hepatocyte cell size, and promoted growth of the liver. These results suggest that cyclin D1 is a key mediator of increased protein synthesis, cell growth, and proliferation downstream of target of rapamycin in mitogen-stimulated hepatocytes.


Experimental Cell Research | 2008

ECTOPIC EXPRESSION OF CYCLIN D3 CORRECTS DIFFERENTIATION OF DM1 MYOBLASTS THROUGH ACTIVATION OF RNA CUG-BINDING PROTEIN, CUGBP1

Elizabeth Salisbury; Keiko Sakai; Benedikt Schoser; Claudia Huichalaf; Christiane Schneider-Gold; Heather Nguyen; Gou Li Wang; Jeffrey H. Albrecht; Lubov Timchenko

Differentiation of myocytes is impaired in patients with myotonic dystrophy type 1, DM1. CUG repeat binding protein, CUGBP1, is a key regulator of translation of proteins that are involved in muscle development and differentiation. In this paper, we present evidence that RNA-binding activity of CUGBP1 and its interactions with initiation translation complex eIF2 are differentially regulated during myogenesis by specific phosphorylation and that this regulation is altered in DM1. In normal myoblasts, Akt kinase phosphorylates CUGBP1 at Ser28 and increases interactions of CUGBP1 with cyclin D1 mRNA. During differentiation, CUGBP1 is phosphorylated by cyclinD3-cdk4/6 at Ser302, which increases CUGBP1 binding with p21 and C/EBPbeta mRNAs. While cyclin D3 and cdk4 are elevated in normal myotubes; DM1 differentiating cells do not increase these proteins. In normal myotubes, CUGBP1 interacts with cyclin D3/cdk4/6 and eIF2; however, interactions of CUGBP1 with eIF2 are reduced in DM1 differentiating cells and correlate with impaired muscle differentiation in DM1. Ectopic expression of cyclin D3 in DM1 cells increases the CUGBP1-eIF2 complex, corrects expression of differentiation markers, myogenin and desmin, and enhances fusion of DM1 myoblasts. Thus, normalization of cyclin D3 might be a therapeutic approach to correct differentiation of skeletal muscle in DM1 patients.


The EMBO Journal | 2002

C/EBPα triggers proteasome‐dependent degradation of cdk4 during growth arrest

Hongmei Wang; Triona Goode; Polina Iakova; Jeffrey H. Albrecht; Nikolai A. Timchenko

CCAAT/enhancer binding protein alpha (C/EBPα) causes growth arrest via direct interaction with the cyclin‐dependent kinases cdk2 and cdk4. In this paper, we present evidence showing that C/EBPα enhances a proteasome‐dependent degradation of cdk4 during growth arrest in liver of newborn mice and in cultured cells. Overexpression of C/EBPα in several biological systems leads to a reduction of cdk4 protein levels, but not mRNA levels. Experiments with several tissue culture models reveal that C/EBPα enhances the formation of cdk4–ubiquitin conjugates and induces degradation of cdk4 through a proteasome‐dependent pathway. As a result, the half‐life of cdk4 is shorter and protein levels of cdk4 are reduced in cells expressing C/EBPα. Gel filtration analysis of cdk4 complexes shows that a chaperone complex cdk4–cdc37–Hsp90, which protects cdk4 from degradation, is abundant in proliferating livers that lack C/EBPα, but this complex is weak or undetectable in livers expressing C/EBPα. Our studies show that C/EBPα disrupts the cdk4–cdc37–Hsp90 complex via direct interaction with cdk4 and reduces protein levels of cdk4 by increasing proteasome‐dependent degradation of cdk4.


Journal of Biological Chemistry | 2006

Age-specific CUGBP1-eIF2 Complex Increases Translation of CCAAT/Enhancer-binding Protein β in Old Liver

Lubov Timchenko; Elizabeth Salisbury; Guo Li Wang; Heather Nguyen; Jeffrey H. Albrecht; John W. B. Hershey; Nikolai A. Timchenko

The RNA-binding protein CUGBP1 regulates translation of proteins in a variety of biological processes. In this study, we show that aging liver increases CUGBP1 translational activities by induction of a high molecular weight protein-protein complex of CUGBP1. The complex contains CUGBP1, subunits α, β, and γ of the initiation translation factor eIF2, and four proteins of the endoplasmic reticulum, eR90, CRT, eR60, and Grp78. The induction of the CUGBP1-eIF2 complex in old livers is associated with the elevation of protein levels of CUGBP1 and with the hyper-phosphorylation of CUGBP1 by a cyclin D3-cdk4 kinase, activity of which is increased with age. We have examined the role of the elevation of CUGBP1 and the role of cyclin D3-cdk4-mediated phosphorylation of CUGBP1 in the formation of the CUGBP1-eIF2 complex by using CUGBP1 transgenic mice and young animals expressing high levels of cyclin D3 after injection with cyclin D3 plasmid. These studies showed that both the increased levels of CUGBP1 and cdk4-mediated hyper-phosphorylation of CUGBP1 are involved in the age-associated induction of the CUGBP1-eIF2 complex. The CUGBP1-eIF2 complex is bound to C/EBPβ mRNA in the liver of old animals, and this binding correlates with the increased amounts of liver-enriched activator protein and liver-enriched inhibitory protein. Consistent with these observations, the purified CUGBP1-eIF2 complex binds to the 5′ region of C/EBPβ mRNA and significantly increases translation of the three isoforms of C/EBPβ in a cell-free translation system, in cultured cells, and in the liver. Thus, these studies demonstrated that age-mediated induction of the CUGBP1-eIF2 complex changes translation of C/EBPβ in old livers.


American Journal of Physiology-gastrointestinal and Liver Physiology | 1998

Effects of chronic ethanol consumption on cytokine regulation of liver regeneration

Shi Qi Yang; Hui Zhi Lin; Ming Yin; Jeffrey H. Albrecht; Anna Mae Diehl

Ethanol ingestion may interrupt the proregenerative signal transduction that is initiated by injury-related cytokines such as tumor necrosis factor (TNF)-α and TNF-α- inducible cytokines including interleukin (IL)-6. To test this theory, liver regeneration, TNF-α and IL-6 expression, and cytokine-regulated prereplicative events were compared in ethanol-fed rats and isocalorically fed controls after 70% partial hepatectomy (PH). Ethanol feeding inhibits hepatocyte replication and recovery of liver mass after PH but generally promotes induction of both cytokines in the liver and extrahepatic tissues (i.e., white adipose tissue). Cytokine-regulated events that occur early in the prereplicative period are influenced differentially. TNF-α-dependent increases in hepatic nuclear factor-κB (NF-κB) p50 and p65 expression and DNA binding activity are prevented, whereas IL-6-dependent inductions of hepatic Stat-3 phosphorylation and DNA binding activity occur normally. In contrast, events (e.g., induction of cyclin D1, cdk-1, cyclin D3, and p53 mRNA) that occur at the end of the prereplicative period are uniformly inhibited. These findings indicate that chronic ethanol ingestion arrests the regenerative process during the prereplicative period and demonstrate that increased TNF-α, IL-6 and Stat-3 are not sufficient to assure hepatocyte proliferation after PH.


Hepatology | 2011

Genomewide microRNA down-regulation as a negative feedback mechanism in the early phases of liver regeneration.

Jingmin Shu; Betsy T. Kren; Zhilian Xia; Phillip Y.-P. Wong; Lihua Li; Eric A. Hanse; Michael Xinan Min; Bingshan Li; Jeffrey H. Albrecht; Yan Zeng; Subbaya Subramanian; Clifford J. Steer

The liver is one of the few organs that have the capacity to regenerate in response to injury. We carried out genomewide microRNA (miRNA) microarray studies during liver regeneration in rats after 70% partial hepatectomy (PH) at early and mid time points to more thoroughly understand their role. At 3, 12, and 18 hours post‐PH ∼40% of the miRNAs tested were up‐regulated. Conversely, at 24 hours post‐PH, ∼70% of miRNAs were down‐regulated. Furthermore, we established that the genomewide down‐regulation of miRNA expression at 24 hours was also correlated with decreased expression of genes, such as Rnasen, Dgcr8, Dicer, Tarbp2, and Prkra, associated with miRNA biogenesis. To determine whether a potential negative feedback loop between miRNAs and their regulatory genes exists, 11 candidate miRNAs predicted to target the above‐mentioned genes were examined and found to be up‐regulated at 3 hours post‐PH. Using reporter and functional assays, we determined that expression of these miRNA‐processing genes could be regulated by a subset of miRNAs and that some miRNAs could target multiple miRNA biogenesis genes simultaneously. We also demonstrated that overexpression of these miRNAs inhibited cell proliferation and modulated cell cycle in both Huh‐7 human hepatoma cells and primary rat hepatocytes. From these observations, we postulated that selective up‐regulation of miRNAs in the early phase after PH was involved in the priming and commitment to liver regeneration, whereas the subsequent genomewide down‐regulation of miRNAs was required for efficient recovery of liver cell mass. Conclusion: Our data suggest that miRNA changes are regulated by negative feedback loops between miRNAs and their regulatory genes that may play an important role in the steady‐state regulation of liver regeneration. (HEPATOLOGY 2011;)


Molecular and Cellular Biology | 2006

Cyclin D3 Maintains Growth-Inhibitory Activity of C/EBPα by Stabilizing C/EBPα-cdk2 and C/EBPα-Brm Complexes

Guo Li Wang; Xiurong Shi; Elizabeth Salisbury; Yuxiang Sun; Jeffrey H. Albrecht; Roy G. Smith; Nikolai A. Timchenko

ABSTRACT C/EBPα arrests proliferation of young livers by inhibition of cdk2. In old mice, C/EBPα inhibits growth by repression of E2F-dependent promoters through the C/EBPα-Brm complex. In this paper, we show that cyclin D3-cdk4/cdk6 supports the ability of C/EBPα to inhibit liver proliferation in both age groups. Although cyclin D3-cdk4/cdk6 kinases are involved in the promotion of growth, they are expressed in terminally differentiated cells, suggesting that they have additional functions in these settings. We demonstrate that C/EBPα represents a target for phosphorylation by cyclin D3-cdk4/cdk6 complexes in differentiated liver cells and in differentiated adipocytes. Cyclin D3-cdk4/cdk6 specifically phosphorylate C/EBPα at Ser193 in vitro and in the liver and support growth-inhibitory C/EBPα-cdk2 and C/EBPα-Brm complexes. We found that cyclin D3 is increased in old livers and activates cdk4/cdk6, resulting in stabilization of the C/EBPα-Brm complex. Old livers fail to reduce the activity of cyclin D3-cdk4/cdk6 after partial hepatectomy, leading to high levels of C/EBPα-Brm complexes after partial hepatectomy, which correlate with weak proliferation. We examined the role of cyclin D3 in the stabilization of C/EBPα-cdk2 and C/EBPα-Brm by using 3T3-L1 differentiated cells. In these cells, cyclin D3 is increased during differentiation and phosphorylates C/EBPα at Ser193, leading to the formation of growth-inhibitory C/EBPα-cdk2 and C/EBPα-Brm complexes. The inhibition of cyclin D3 blocks the formation of these complexes. Thus, these studies provide a new function of cyclin D3, which is to support the growth-inhibitory activity of C/EBPα.


Journal of Clinical Investigation | 2011

S6 kinase 1 is required for rapamycin-sensitive liver proliferation after mouse hepatectomy

Catherine Espeillac; Claudia Mitchell; Séverine Celton-Morizur; Céline Chauvin; Vonda Koka; Cynthia Gillet; Jeffrey H. Albrecht; Chantal Desdouets; Mario Pende

Rapamycin is an antibiotic inhibiting eukaryotic cell growth and proliferation by acting on target of rapamycin (TOR) kinase. Mammalian TOR (mTOR) is thought to work through 2 independent complexes to regulate cell size and cell replication, and these 2 complexes show differential sensitivity to rapamycin. Here we combine functional genetics and pharmacological treatments to analyze rapamycin-sensitive mTOR substrates that are involved in cell proliferation and tissue regeneration after partial hepatectomy in mice. After hepatectomy, hepatocytes proliferated rapidly, correlating with increased S6 kinase phosphorylation, while treatment with rapamycin derivatives impaired regeneration and blocked S6 kinase activation. In addition, genetic deletion of S6 kinase 1 (S6K1) caused a delay in S phase entry in hepatocytes after hepatectomy. The proliferative defect of S6K1-deficient hepatocytes was cell autonomous, as it was also observed in primary cultures and hepatic overexpression of S6K1-rescued proliferation. We found that S6K1 controlled steady-state levels of cyclin D1 (Ccnd1) mRNA in liver, and cyclin D1 expression was required to promote hepatocyte cell cycle. Notably, in vivo overexpression of cyclin D1 was sufficient to restore the proliferative capacity of S6K-null livers. The identification of an S6K1-dependent mechanism participating in cell proliferation in vivo may be relevant for cancer cells displaying high mTOR complex 1 activity and cyclin D1 accumulation.

Collaboration


Dive into the Jeffrey H. Albrecht's collaboration.

Top Co-Authors

Avatar

Christopher J. Nelsen

Hennepin County Medical Center

View shared research outputs
Top Co-Authors

Avatar

Eric A. Hanse

Hennepin County Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lisa K. Mullany

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nikolai A. Timchenko

Hennepin County Medical Center

View shared research outputs
Top Co-Authors

Avatar

Andrew Conrad

University of California

View shared research outputs
Top Co-Authors

Avatar

David G. Rickheim

Hennepin County Medical Center

View shared research outputs
Top Co-Authors

Avatar

Melissa M. Goggin

Hennepin County Medical Center

View shared research outputs
Top Co-Authors

Avatar

Scott J. Cotler

Loyola University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge