Network


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

Hotspot


Dive into the research topics where Razqallah Hakem is active.

Publication


Featured researches published by Razqallah Hakem.


Cell | 1998

Differential requirement for caspase 9 in apoptotic pathways in vivo.

Razqallah Hakem; Anne Hakem; Gordon S. Duncan; Jeffrey T. Henderson; Minna Woo; Maria S. Soengas; Andrew Elia; José Luis de la Pompa; David Kagi; Wilson Khoo; Julia Potter; Ritsuko Yoshida; Stephen Kaufman; Scott W. Lowe; Josef M. Penninger; Tak W. Mak

Mutation of Caspase 9 (Casp9) results in embryonic lethality and defective brain development associated with decreased apoptosis. Casp9-/- embryonic stem cells and embryonic fibroblasts are resistant to several apoptotic stimuli, including UV and gamma irradiation. Casp9-/- thymocytes are also resistant to dexamethasone- and gamma irradiation-induced apoptosis, but are surprisingly sensitive to apoptosis induced by UV irradiation or anti-CD95. Resistance to apoptosis is accompanied by retention of the mitochondrial membrane potential in mutant cells. In addition, cytochrome c is translocated to the cytosol of Casp9-/- ES cells upon UV stimulation, suggesting that Casp9 acts downstream of cytochrome c. Caspase processing is inhibited in Casp9-/- ES cells but not in thymocytes or splenocytes. Comparison of the requirement for Casp9 and Casp3 in different apoptotic settings indicates the existence of at least four different apoptotic pathways in mammalian cells.


Nature | 2001

Essential role of the mitochondrial apoptosis-inducing factor in programmed cell death

Nicholas Joza; Santos A. Susin; Eric Daugas; William L. Stanford; Sarah K. Cho; Carol Y. J. Li; Takehiko Sasaki; Andrew J. Elia; H.-Y. Mary Cheng; Luigi Ravagnan; Karine F. Ferri; Naoufal Zamzami; Andrew Wakeham; Razqallah Hakem; Hiroki Yoshida; Young-Yun Kong; Tak W. Mak; Juan Carlos Zúñiga-Pflücker; Guido Kroemer; Josef M. Penninger

Programmed cell death is a fundamental requirement for embryogenesis, organ metamorphosis and tissue homeostasis. In mammals, release of mitochondrial cytochrome c leads to the cytosolic assembly of the apoptosome—a caspase activation complex involving Apaf1 and caspase-9 that induces hallmarks of apoptosis. There are, however, mitochondrially regulated cell death pathways that are independent of Apaf1/caspase-9. We have previously cloned a molecule associated with programmed cell death called apoptosis-inducing factor (AIF). Like cytochrome c, AIF is localized to mitochondria and released in response to death stimuli. Here we show that genetic inactivation of AIF renders embryonic stem cells resistant to cell death after serum deprivation. Moreover, AIF is essential for programmed cell death during cavitation of embryoid bodies—the very first wave of cell death indispensable for mouse morphogenesis. AIF-dependent cell death displays structural features of apoptosis, and can be genetically uncoupled from Apaf1 and caspase-9 expression. Our data provide genetic evidence for a caspase-independent pathway of programmed cell death that controls early morphogenesis.


Cell | 1998

Apaf1 is required for mitochondrial pathways of apoptosis and brain development.

Hiroki Yoshida; Young-Yun Kong; Ritsuko Yoshida; Andrew J. Elia; Anne Hakem; Razqallah Hakem; Josef M. Penninger; Tak W. Mak

Apoptosis is essential for the precise regulation of cellular homeostasis and development. The role in vivo of Apaf1, a mammalian homolog of C. elegans CED-4, was investigated in gene-targeted Apaf1-/- mice. Apaf1-deficient mice exhibited reduced apoptosis in the brain and striking craniofacial abnormalities with hyperproliferation of neuronal cells. Apaf1-deficient cells were resistant to a variety of apoptotic stimuli, and the processing of Caspases 2, 3, and 8 was impaired. However, both Apaf1-/- thymocytes and activated T lymphocytes were sensitive to Fas-induced killing, showing that Fas-mediated apoptosis in these cells is independent of Apaf1. These data indicate that Apaf1 plays a central role in the common events of mitochondria-dependent apoptosis in most death pathways and that this role is critical for normal development.


Nature | 2011

RIP3 mediates the embryonic lethality of caspase-8-deficient mice

William J. Kaiser; Jason W. Upton; Alyssa B. Long; Devon Livingston-Rosanoff; Lisa P. Daley-Bauer; Razqallah Hakem; Tamara Caspary; Edward S. Mocarski

Apoptosis and necroptosis are complementary pathways controlled by common signalling adaptors, kinases and proteases; among these, caspase-8 (Casp8) is critical for death receptor-induced apoptosis. This caspase has also been implicated in non-apoptotic pathways that regulate Fas-associated via death domain (FADD)-dependent signalling and other less defined biological processes as diverse as innate immune signalling and myeloid or lymphoid differentiation patterns. Casp8 suppresses RIP3–RIP1 (also known as RIPK3–RIPK1) kinase complex-dependent necroptosis that follows death receptor activation as well as a RIP3-dependent, RIP1-independent necrotic pathway that has emerged as a host defence mechanism against murine cytomegalovirus. Disruption of Casp8 expression leads to embryonic lethality in mice between embryonic days 10.5 and 11.5 (ref. 7). Thus, Casp8 may naturally hold alternative RIP3-dependent death pathways in check in addition to promoting apoptosis. We find that RIP3 is responsible for the mid-gestational death of Casp8-deficient embryos. Remarkably, Casp8−/−Rip3−/− double mutant mice are viable and mature into fertile adults with a full immune complement of myeloid and lymphoid cell types. These mice seem immunocompetent but develop lymphadenopathy by four months of age marked by accumulation of abnormal T cells in the periphery, a phenotype reminiscent of mice with Fas-deficiency (lpr/lpr; also known as Fas). Thus, Casp8 contributes to homeostatic control in the adult immune system; however, RIP3 and Casp8 are together completely dispensable for mammalian development.


Cell | 2003

Pirh2, a p53-Induced Ubiquitin-Protein Ligase, Promotes p53 Degradation

Roger P. Leng; Yunping Lin; Weili Ma; Hong Wu; Bénédicte Lemmers; Stephen W. Chung; John M. Parant; Guillermina Lozano; Razqallah Hakem; Samuel Benchimol

The p53 tumor suppressor exerts anti-proliferative effects in response to various types of stress including DNA damage and abnormal proliferative signals. Tight regulation of p53 is essential for maintaining normal cell growth and this occurs primarily through posttranslational modifications of p53. Here, we describe Pirh2, a gene regulated by p53 that encodes a RING-H2 domain-containing protein with intrinsic ubiquitin-protein ligase activity. Pirh2 physically interacts with p53 and promotes ubiquitination of p53 independently of Mdm2. Expression of Pirh2 decreases the level of p53 protein and abrogation of endogenous Pirh2 expression increases the level of p53. Furthermore, Pirh2 represses p53 functions including p53-dependent transactivation and growth inhibition. We propose that Pirh2 is involved in the negative regulation of p53 function through physical interaction and ubiquitin-mediated proteolysis. Hence, Pirh2, like Mdm2, participates in an autoregulatory feedback loop that controls p53 function.


Cell | 1996

The Tumor Suppressor Gene Brca1 Is Required for Embryonic Cellular Proliferation in the Mouse

Razqallah Hakem; José Luis de la Pompa; Christian Sirard; Rong Mo; Minna Woo; Anne Hakem; Andrew Wakeham; Julia Potter; Armin H. Reitmair; Filio Billia; Eduardo Firpo; Chi Chung Hui; Jim Roberts; Janet Rossant; Tak W. Mak

Mutations of the BRCA1 gone in humans are associated with predisposition to breast and ovarian cancers. We show here that Brca1+/- mice are normal and fertile and lack tumors by age eleven months. Homozygous Brca1(5-6) mutant mice die before day 7.5 of embryogenesis. Mutant embryos are poorly developed, with no evidence of mesoderm formation. The extraembryonic region is abnormal, but aggregation with wild-type tetraploid embryos does not rescue the lethality. In vivo, mutant embryos do not exhibit increased apoptosis but show reduced cell proliferation accompanied by decreased expression of cyclin E and mdm-2, a regulator of p53 activity. The expression of cyclin-dependent kinase inhibitor p21 is dramatically increased in the mutant embryos. Buttressing these in vivo observations is the fact that mutant blastocyst growth is grossly impaired in vitro. Thus, the death of Brca1(5-6) mutant embryos prior to gastrulation may be due to a failure of the proliferative burst required for the development of the different germ layers.


Molecular and Cellular Biology | 2000

BNIP3 and Genetic Control of Necrosis-Like Cell Death through the Mitochondrial Permeability Transition Pore

C. Vande Velde; Don Dubik; J. Alimonti; T. Brown; S. Israels; Razqallah Hakem; Arnold H. Greenberg

ABSTRACT Many apoptotic signaling pathways are directed to mitochondria, where they initiate the release of apoptogenic proteins and open the proposed mitochondrial permeability transition (PT) pore that ultimately results in the activation of the caspase proteases responsible for cell disassembly. BNIP3 (formerly NIP3) is a member of the Bcl-2 family that is expressed in mitochondria and induces apoptosis without a functional BH3 domain. We report that endogenous BNIP3 is loosely associated with mitochondrial membrane in normal tissue but fully integrates into the mitochondrial outer membrane with the N terminus in the cytoplasm and the C terminus in the membrane during induction of cell death. Surprisingly, BNIP3-mediated cell death is independent of Apaf-1, caspase activation, cytochrome c release, and nuclear translocation of apoptosis-inducing factor. However, cells transfected with BNIP3 exhibit early plasma membrane permeability, mitochondrial damage, extensive cytoplasmic vacuolation, and mitochondrial autophagy, yielding a morphotype that is typical of necrosis. These changes were accompanied by rapid and profound mitochondrial dysfunction characterized by opening of the mitochondrial PT pore, proton electrochemical gradient (Δψm) suppression, and increased reactive oxygen species production. The PT pore inhibitors cyclosporin A and bongkrekic acid blocked mitochondrial dysregulation and cell death. We propose that BNIP3 is a gene that mediates a necrosis-like cell death through PT pore opening and mitochondrial dysfunction.


The EMBO Journal | 2008

DNA‐damage repair; the good, the bad, and the ugly

Razqallah Hakem

Organisms have developed several DNA‐repair pathways as well as DNA‐damage checkpoints to cope with the frequent challenge of endogenous and exogenous DNA insults. In the absence or impairment of such repair or checkpoint mechanisms, the genomic integrity of the organism is often compromised. This review will focus on the functional consequences of impaired DNA‐repair pathways. Although each pathway is addressed individually, it is essential to note that cross talk exists between repair pathways, and that there are instances in which a DNA‐repair protein is involved in more than one pathway. It is also important to integrate DNA‐repair process with DNA‐damage checkpoints and cell survival, to gain a better understanding of the consequences of compromised DNA repair at both cellular and organismic levels. Functional consequences associated with impaired DNA repair include embryonic lethality, shortened life span, rapid ageing, impaired growth, and a variety of syndromes, including a pronounced manifestation of cancer.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Dysregulation of the mevalonate pathway promotes transformation

James W. Clendening; Aleks Pandyra; Paul C. Boutros; Samah El Ghamrasni; Fereshteh Khosravi; Grace A. Trentin; Anna Martirosyan; Anne Hakem; Razqallah Hakem; Igor Jurisica; Linda Z. Penn

The importance of cancer metabolism has been appreciated for many years, but the intricacies of how metabolic pathways interconnect with oncogenic signaling are not fully understood. With a clear understanding of how metabolism contributes to tumorigenesis, we will be better able to integrate the targeting of these fundamental biochemical pathways into patient care. The mevalonate (MVA) pathway, paced by its rate-limiting enzyme, hydroxymethylglutaryl coenzyme A reductase (HMGCR), is required for the generation of several fundamental end-products including cholesterol and isoprenoids. Despite years of extensive research from the perspective of cardiovascular disease, the contribution of a dysregulated MVA pathway to human cancer remains largely unexplored. We address this issue directly by showing that dysregulation of the MVA pathway, achieved by ectopic expression of either full-length HMGCR or its novel splice variant, promotes transformation. Ectopic HMGCR accentuates growth of transformed and nontransformed cells under anchorage-independent conditions or as xenografts in immunocompromised mice and, importantly, cooperates with RAS to drive the transformation of primary mouse embryonic fibroblasts cells. We further explore whether the MVA pathway may play a role in the etiology of human cancers and show that high mRNA levels of HMGCR and additional MVA pathway genes correlate with poor prognosis in a meta-analysis of six microarray datasets of primary breast cancer. Taken together, our results suggest that HMGCR is a candidate metabolic oncogene and provide a molecular rationale for further exploring the statin family of HMGCR inhibitors as anticancer agents.


The EMBO Journal | 2004

Lats2/Kpm is required for embryonic development, proliferation control and genomic integrity

John McPherson; Laura Tamblyn; Andrew Elia; Eva Migon; Amro Shehabeldin; Elzbieta Matysiak-Zablocki; Bénédicte Lemmers; Leonardo Salmena; Anne Hakem; Jason Fish; Farah Kassam; Jeremy A. Squire; Benoit G. Bruneau; M. Prakash Hande; Razqallah Hakem

The Drosophila melanogaster warts/lats tumour suppressor has two mammalian counterparts LATS1/Warts‐1 and LATS2/Kpm. Here, we show that mammalian Lats orthologues exhibit distinct expression profiles according to germ cell layer origin. Lats2−/− embryos show overgrowth in restricted tissues of mesodermal lineage; however, lethality ultimately ensues on or before embryonic day 12.5 preceded by defective proliferation. Lats2−/− mouse embryonic fibroblasts (MEFs) acquire growth advantages and display a profound defect in contact inhibition of growth, yet exhibit defective cytokinesis. Lats2−/− embryos and MEFs display centrosome amplification and genomic instability. Lats2 localizes to centrosomes and overexpression of Lats2 suppresses centrosome overduplication induced in wild‐type MEFs and reverses centrosome amplification inherent in Lats2−/− MEFs. These findings indicate an essential role of Lats2 in the integrity of processes that govern centrosome duplication, maintenance of mitotic fidelity and genomic stability.

Collaboration


Dive into the Razqallah Hakem's collaboration.

Top Co-Authors

Avatar

Anne Hakem

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Tak W. Mak

University Health Network

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Minna Woo

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Otto Sanchez

University of Ontario Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Bénédicte Lemmers

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Elzbieta Matysiak-Zablocki

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

M. Prakash Hande

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Renato Cardoso

University Health Network

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge