Network


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

Hotspot


Dive into the research topics where Jill M. Lahti is active.

Publication


Featured researches published by Jill M. Lahti.


Nature Medicine | 2000

Caspase 8 is deleted or silenced preferentially in childhood neuroblastomas with amplification of MYCN.

Tal Teitz; Tie Wei; Marcus B. Valentine; Vanin Ef; Jose Grenet; Virginia Valentine; Behm Fg; Look At; Jill M. Lahti; Vincent J. Kidd

Caspase 8 is a cysteine protease regulated in both a death-receptor-dependent and -independent manner during apoptosis. Here, we report that the gene for caspase 8 is frequently inactivated in neuroblastoma, a childhood tumor of the peripheral nervous system. The gene is silenced through DNA methylation as well as through gene deletion. Complete inactivation of CASP8 occurred almost exclusively in neuroblastomas with amplification of the oncogene MYCN. Caspase 8-null neuroblastoma cells were resistant to death receptor- and doxorubicin-mediated apoptosis, deficits that were corrected by programmed expression of the enzyme. Thus, caspase 8 acts as a tumor suppressor in neuroblastomas with amplification of MYCN.


Journal of Biological Chemistry | 2002

ERK activation mediates cell cycle arrest and apoptosis after DNA damage independently of p53.

Damu Tang; Dongcheng Wu; Atsushi Hirao; Jill M. Lahti; Lieqi Liu; Brie Mazza; Vincent J. Kidd; Tak W. Mak; Alistair J. Ingram

In response to DNA damage, ataxia-telangiectasia mutant and ataxia-telangiectasia and Rad-3 activate p53, resulting in either cell cycle arrest or apoptosis. We report here that DNA damage stimuli, including etoposide (ETOP), adriamycin (ADR), ionizing irradiation (IR), and ultraviolet irradiation (UV) activate ERK1/2 (ERK) mitogen-activated protein kinase in primary (MEF and IMR90), immortalized (NIH3T3) and transformed (MCF-7) cells. ERK activation in response to ETOP was abolished in ATM−/− fibroblasts (GM05823) and was independent of p53. The MEK1 inhibitor PD98059 prevented ERK activation but not p53 stabilization. Maximal ERK activation in response to DNA damage was not attenuated in MEFp53−/−. However, ERK activation contributes to either cell cycle arrest or apoptosis in response to low or high intensity DNA insults, respectively. Inhibition of ERK activation by PD98059 or U0126 attenuated p21CIP1 induction, resulting in partial release of the G2/M cell cycle arrest induced by ETOP. Furthermore, PD98059 or U0126 also strongly attenuated apoptosis induced by high dose ETOP, ADR, or UV. Conversely, enforced activation of ERK by overexpression of MEK-1/Q56P sensitized cells to DNA damage-induced apoptosis. Taken together, these results indicate that DNA damage activates parallel ERK and p53 pathways in an ATM-dependent manner. These pathways might function cooperatively in cell cycle arrest and apoptosis.


Nature | 2006

Potentiation of neuroblastoma metastasis by loss of caspase-8.

Dwayne G. Stupack; Tal Teitz; Matthew D. Potter; David Mikolon; Peter J. Houghton; Vincent J. Kidd; Jill M. Lahti; David A. Cheresh

Neuroblastoma, the most common paediatric solid tumour, arises from defective neural crest cells. Genetic alterations occur frequently in the most aggressive neuroblastomas. In particular, deletion or suppression of the proapoptotic enzyme caspase-8 is common in malignant, disseminated disease, although the effect of this loss on disease progression is unclear. Here we show that suppression of caspase-8 expression occurs during the establishment of neuroblastoma metastases in vivo, and that reconstitution of caspase-8 expression in deficient neuroblastoma cells suppressed their metastases. Caspase-8 status was not a predictor of primary tumour growth; rather, caspase-8 selectively potentiated apoptosis in neuroblastoma cells invading the collagenous stroma at the tumour margin. Apoptosis was initiated by unligated integrins by means of a process known as integrin-mediated death. Loss of caspase-8 or integrin rendered these cells refractory to integrin-mediated death, allowed cellular survival in the stromal microenvironment, and promoted metastases. These findings define caspase-8 as a metastasis suppressor gene that, together with integrins, regulates the survival and invasive capacity of neuroblastoma cells.


Molecular and Cellular Biology | 1995

PITSLRE protein kinase activity is associated with apoptosis.

Jill M. Lahti; Jialing Xiang; Lucie S. Heath; Dario Campana; Vincent J. Kidd

Minimal ectopic expression of a 58-kDa protein kinase (PITSLRE beta 1), distantly related to members of the cdc2 gene family, induces telophase delay, abnormal chromosome segregation, and decreased growth rates in Chinese hamster ovary cells. Here we show that this decrease in cell growth rate is due to apoptosis. Apoptosis is also induced by ectopic expression of an amino-terminal deletion mutant containing the catalytic and C-terminal domains of PITSLRE beta 1 but not by other mutants lacking histone H1 kinase activity or by other members of the cdc2 gene family. However, unlike the wild-type PITSLRE beta 1 over-expressors, ectopic expression of the N-terminal PITSLRE beta 1 mutant does not result in telophase delay or abnormal chromosome segregation. These results suggested that the function of this protein kinase could be linked to apoptotic signaling. To test this hypothesis, we examined levels of PITSLRE mRNA, steady-state protein, and enzyme activity in human T cells undergoing apoptosis after activation with the anti-Fas monoclonal antibody (MAb). All were substantially elevated shortly after Fas MAb treatment. In addition to new transcription and translation, proteolysis contributed to the increased steady-state levels of a novel 50-kDa PITSLRE protein, as suggested by the diminution of larger PITSLRE isoforms observed in the same cells. Indeed, treatment of the Fas-activated T cells with a serine protease inhibitor prevented apoptotic death and led to the accumulation of larger, less active PITSLRE kinase isoforms but not the enzymatically active 50-kDa PITSLRE isoform. Finally, induction of apoptosis by glucocorticoids in the same cell line, as well as by Fas MAb treatment of another T-cell line, led to a similar induction of 50-kDa PITSLRE protein levels over time. These findings suggest that (i) PITSLRE kinase(s) may lie within apoptotic signaling pathway(s), (ii) serine protease activation may be an early event in Fas-activated apoptosis of human T cells, and (iii) some PITSLRE kinase isoforms may be targets of apoptotic proteases.


Nature Cell Biology | 2009

Cyclin-dependent kinases: a family portrait.

Marcos Malumbres; Ed Harlow; Tim Hunt; Tony Hunter; Jill M. Lahti; Gerard Manning; David O. Morgan; Li-Huei Tsai; Debra J. Wolgemuth

To the Editor, Cyclin-dependent kinases (CDKs) are protein kinases involved in critical cellular processes, such as cell cycle or transcription, whose activity requires association with specific cyclin subunits. Based on sequence similarity, the human genome contains 21 genes encoding CDKs and five additional genes encoding a more distant group of proteins known as CDK-like (CDKL) kinases. The current nomenclature for CDK proteins includes 11 classical CDKs (CDK1–11), two newly proposed family members (CDK12 and 13) and additional proteins whose names are based on the presence of a cyclin-binding element (PFTAIRE and PCTAIRE proteins) or simply based on a sequence relationship with the original CDKs, such as CDC2-like kinases (CDC2L) or cell cycle-related kinases (CCRK)


Journal of Biological Chemistry | 1999

Cycloheximide-induced T-cell Death Is Mediated by a Fas-associated Death Domain-dependent Mechanism

Damu Tang; Jill M. Lahti; Jose Grenet; Vincent J. Kidd

Cycloheximide (CHX) can contribute to apoptotic processes, either in conjunction with another agent (e.g. tumor necrosis factor-α) or on its own. However, the basis of this CHX-induced apoptosis has not been clearly established. In this study, the molecular mechanisms of CHX-induced cell death were examined in two different human T-cell lines. In T-cells undergoing CHX-induced apoptosis (Jurkat), but not in T-cells resistant to the effects of CHX (CEM C7), caspase-8 and caspase-3 were activated. However, the Fas ligand was not expressed in Jurkat cells either before or after treatment with CHX, suggesting that the activation of these caspases does not involve the Fas receptor. To determine whether CHX-induced apoptosis was mediated by a Fas-associated death domain (FADD)-dependent mechanism, a FADD-DN protein was expressed in cells prior to CHX treatment. Its expression effectively inhibited CHX-induced cell death, suggesting that CHX-mediated apoptosis primarily involves a FADD-dependent mechanism. Since CHX treatment did not result in the induction of Fas or FasL, and neutralizing anti-Fas and anti-tumor necrosis factor receptor-1 antibodies did not block CHX-mediated apoptosis, these results may also indicate that FADD functions in a receptor-independent manner. Surprisingly, death effector filaments containing FADD and caspase-8 were observed during CHX treatment of Jurkat, Jurkat-FADD-DN, and CEM C7 cells, suggesting that their formation may be necessary, but not sufficient, for cell death.


Journal of Molecular Medicine | 2001

Aggressive childhood neuroblastomas do not express caspase-8: an important component of programmed cell death

Tal Teitz; Jill M. Lahti; Vincent J. Kidd

Neuroblastomas that overexpress N-Myc due to amplification of the MYCN oncogene are aggressive tumors that become very resistant to treatment by chemotherapy and irradiation. to identify tumor suppressor genes in this group of neuroblastomas we analyzed the expression and function of both apoptosis-related cell cycle regulatory genes in cell lines and patient tumor samples. We found that in a high percentage of neuroblastoma cell lines and patient samples with amplified MYCN, caspase-8 mRNA is not expressed. The caspase-8 gene, CASP8, was deleted or silenced by methylation in the neuroblastoma cell lines while methylation of its promoter region was the predominant mechanism for its inactivation in the patient tumor samples. Reintroduction of caspase-8 into the neuroblastoma cell lines resensitized these cells to drug-induced and survival factor dependent apoptosis. Subsequently others have also shown that caspase-8 is silenced by methylation in neuroblastoma and peripheral neural ectodermal tumors, and that the caspase-9 regulator Apaf-1 is silenced by methylation in melanoma cell lines and patient samples. We conclude that caspase-8 acts as a tumor suppressor gene in neuroblastomas, that its silencing provides a permissive environment for MYCN gene amplification once the tumors are treated with chemotherapeutic drugs/irradiation, and that expression of this gene in these tumor cells may be of clinical benefit. We also discuss the possible significance of the neural crest cell progenitor cell origin and the silencing of important apoptotic regulators via methylation in both neuroblastoma and melanoma tumors.


Cancer Research | 2006

Caspase-8 promotes cell motility and calpain activity under nonapoptotic conditions.

Brooke M. Helfer; Bernard C. Boswell; Darren Finlay; Angel Cipres; Kristiina Vuori; Tae Bong Kang; David Wallach; Andrea Dorfleutner; Jill M. Lahti; Daniel C. Flynn; Steven M. Frisch

Significant caspase-8 activity has been found in normal and certain tumor cells, suggesting that caspase-8 possesses an alternative, nonapoptotic function that may contribute to tumor progression. In this article, we report that caspase-8 promotes cell motility. In particular, caspase-8 is required for the optimal activation of calpains, Rac, and lamellipodial assembly. This represents a novel nonapoptotic function of caspase-8 acting at the intersection of the caspase-8 and calpain proteolytic pathways to coordinate cell death versus cell motility signaling.


Molecular and Cellular Biology | 2003

ASAP, a novel protein complex involved in RNA processing and apoptosis.

Christian Schwerk; Jayendra Prasad; Kurt Degenhardt; Hediye Erdjument-Bromage; Eileen White; Paul Tempst; Vincent J. Kidd; James L. Manley; Jill M. Lahti; Danny Reinberg

ABSTRACT Different isoforms of a protein complex termed the apoptosis- and splicing-associated protein (ASAP) were isolated from HeLa cell extract. ASAP complexes are composed of the polypeptides SAP18 and RNPS1 and different isoforms of the Acinus protein. While Acinus had previously been implicated in apoptosis and was recently identified as a component of the spliceosome, RNPS1 has been described as a general activator of RNA processing. Addition of ASAP isoforms to in vitro splicing reactions inhibits RNA processing mediated by ASF/SF2, by SC35, or by RNPS1. Additionally, microinjection of ASAP complexes into mammalian cells resulted in acceleration of cell death. Importantly, after induction of apoptosis the ASAP complex disassembles. Taken together, our results suggest an important role for the ASAP complexes in linking RNA processing and apoptosis.


Journal of Biological Chemistry | 2008

Characterization of Cyclin L1 and L2 Interactions with CDK11 and Splicing Factors INFLUENCE OF CYCLIN L ISOFORMS ON SPLICE SITE SELECTION

Pascal Loyer; Janeen H. Trembley; Jose Grenet; Adeline Busson; Anne Corlu; Wei Zhao; Mehmet Kocak; Vincent J. Kidd; Jill M. Lahti

Although it has been reported that cyclin L1α and L2α proteins interact with CDK11p110, the nature of the cyclin L transcripts, the formation of complexes between the five cyclin L and the three CDK11 protein isoforms, and the influence of these complexes on splicing have not been thoroughly investigated. Here we report that cyclin L1 and L2 genes generate 14 mRNA variants encoding six cyclin L proteins, one of which has not been described previously. Using cyclin L gene-specific antibodies, we demonstrate expression of multiple endogenous cyclin L proteins in human cell lines and mouse tissues. Moreover, we characterize interactions between CDK11p110, mitosis-specific CDK11p58, and apoptosis-specific CDK11p46 with both cyclin Lα and -β proteins and the co-elution of these proteins following size exclusion chromatography. We further establish that CDK11p110 and associated cyclin Lα/β proteins localize to splicing factor compartments and nucleoplasm and interact with serine/arginine-rich proteins. Importantly, we also determine the effect of CDK11-cyclin L complexes on pre-mRNA splicing. Preincubation of nuclear extracts with purified cyclin Lα and -β isoforms depletes the extract of in vitro splicing activity. Ectopic expression of cyclin L1α, L1β, L2α, or L2β or active CDK11p110 individually enhances intracellular intron splicing activity, whereas expression of CDK11p58/p46 or kinase-dead CDK11p110represses splicing activity. Finally, we demonstrate that expression of cyclins Lα and -β and CDK11p110 strongly and differentially affects alternative splicing in vivo. Together, these data establish that CDK11p110 interacts physically and functionally with cyclin Lα and -β isoforms and SR proteins to regulate splicing.

Collaboration


Dive into the Jill M. Lahti's collaboration.

Top Co-Authors

Avatar

Vincent J. Kidd

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Jose Grenet

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Tal Teitz

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marcus B. Valentine

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Dongli Hu

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Akira Inoue

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Haimin Li

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Judith Hyle

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Kejin Zhu

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge