1
|
The cross-talk of autophagy and apoptosis in breast carcinoma: implications for novel therapies? Biochem J 2022; 479:1581-1608. [PMID: 35904454 DOI: 10.1042/bcj20210676] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 12/12/2022]
Abstract
Breast cancer is still the most common cancer in women worldwide. Resistance to drugs and recurrence of the disease are two leading causes of failure in treatment. For a more efficient treatment of patients, the development of novel therapeutic regimes is needed. Recent studies indicate that modulation of autophagy in concert with apoptosis induction may provide a promising novel strategy in breast cancer treatment. Apoptosis and autophagy are two tightly regulated distinct cellular processes. To maintain tissue homeostasis abnormal cells are disposed largely by means of apoptosis. Autophagy, however, contributes to tissue homeostasis and cell fitness by scavenging of damaged organelles, lipids, proteins, and DNA. Defects in autophagy promote tumorigenesis, whereas upon tumor formation rapidly proliferating cancer cells may rely on autophagy to survive. Given that evasion of apoptosis is one of the characteristic hallmarks of cancer cells, inhibiting autophagy and promoting apoptosis can negatively influence cancer cell survival and increase cell death. Hence, combination of antiautophagic agents with the enhancement of apoptosis may restore apoptosis and provide a therapeutic advantage against breast cancer. In this review, we discuss the cross-talk of autophagy and apoptosis and the diverse facets of autophagy in breast cancer cells leading to novel models for more effective therapeutic strategies.
Collapse
|
2
|
Cell survival and apoptosis signaling as therapeutic target for cancer: marine bioactive compounds. Int J Mol Sci 2013; 14:2334-54. [PMID: 23348928 PMCID: PMC3587990 DOI: 10.3390/ijms14022334] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 01/10/2013] [Accepted: 01/11/2013] [Indexed: 02/06/2023] Open
Abstract
Inhibition of apoptosis leads to activation of cell survival factors (e.g., AKT) causes continuous cell proliferation in cancer. Apoptosis, the major form of cellular suicide, is central to various physiological processes and the maintenance of homeostasis in multicellular organisms. A number of discoveries have clarified the molecular mechanism of apoptosis, thus clarifying the link between apoptosis and cell survival factors, which has a therapeutic outcome. Induction of apoptosis and inhibition of cell survival by anticancer agents has been shown to correlate with tumor response. Cellular damage induces growth arrest and tumor suppression by inducing apoptosis, necrosis and senescence; the mechanism of cell death depends on the magnitude of DNA damage following exposure to various anticancer agents. Apoptosis is mainly regulated by cell survival and proliferating signaling molecules. As a new therapeutic strategy, alternative types of cell death might be exploited to control and eradicate cancer cells. This review discusses the signaling of apoptosis and cell survival, as well as the potential contribution of marine bioactive compounds, suggesting that new therapeutic strategies might follow.
Collapse
|
3
|
Leon-Bollotte L, Subramaniam S, Cauvard O, Plenchette-Colas S, Paul C, Godard C, Martinez-Ruiz A, Legembre P, Jeannin JF, Bettaieb A. S-nitrosylation of the death receptor fas promotes fas ligand-mediated apoptosis in cancer cells. Gastroenterology 2011; 140:2009-18, 2018.e1-4. [PMID: 21354149 DOI: 10.1053/j.gastro.2011.02.053] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 01/27/2011] [Accepted: 02/14/2011] [Indexed: 12/06/2022]
Abstract
BACKGROUND & AIMS Fas belongs to the family of tumor necrosis factor receptors which induce apoptosis. Many cancer cells express Fas but do not undergo Fas-mediated apoptosis. Nitric oxide reverses this resistance by increasing levels of Fas at the plasma membrane. We studied the mechanisms by which NO affects Fas function. METHODS Colon and mammary cancer cell lines were incubated with the NO donor glyceryl trinitrate or lipid A; S-nitrosylation of Fas was monitored using the biotin switch assay. Fas constructs that contained mutations at cysteine residues that prevent S-nitrosylation were used to investigate the involvement of S-nitrosylation in Fas-mediated cell death. Apoptosis was monitored according to morphologic criteria. RESULTS NO induced S-nitrosylation of cysteine residues 199 and 304 in the cytoplasmic part of Fas. In cancer cells that overexpressed wild-type Fas, S-nitrosylation induced Fas recruitment to lipid rafts and sensitized the cells to Fas ligand. In cells that expressed a mutant form of Fas in which cysteine 304 was replaced by valine residue, NO-mediated translocation of Fas to lipid rafts was affected and the death-inducing signal complex and synergistic effect of glyceryl trinitrate-Fas ligand were inhibited significantly. These effects were not observed in cells that expressed Fas with a mutation at cysteine 199. CONCLUSIONS We identified post-translational modifications (S-nitrosylation of cysteine residues 199 and 304) in the cytoplasmic domain of Fas. S-nitrosylation at cysteine 304 promotes redistribution of Fas to lipid rafts, formation of the death-inducing signal complex, and induction of cell death.
Collapse
Affiliation(s)
- Lissbeth Leon-Bollotte
- Ecole Pratique des Hautes Etudes, Tumor Immunology and Immunotherapy Laboratory, Dijon, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Prognostic significance of CD95, P53, and BCL2 expression in extranodal non-Hodgkin’s lymphoma. Ann Hematol 2010; 89:889-96. [DOI: 10.1007/s00277-010-0945-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2009] [Accepted: 03/15/2010] [Indexed: 11/26/2022]
|
5
|
Scholl V, Stefanoff CG, Hassan R, Spector N, Renault IZ. Mutations within the 5' region of FAS/CD95 gene in nodal diffuse large B-cell lymphoma. Leuk Lymphoma 2007; 48:957-63. [PMID: 17487740 DOI: 10.1080/10428190701230858] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
CD95 is a cell-surface receptor that mediates apoptosis. A possible association between CD95 mutations and extranodal diffuse large B-cell lymphomas (DLBCL) has been reported. To further elucidate this question, a mutation analysis within the 5' region and exon 9 of CD95 was performed in a series of 66 DLBCL patients, by polymerase chain reaction, single-strand conformational polymorphism, and sequencing in all cases. Four mutations, all within the 5' region, were detected in three cases of primary nodal DLBCL (6.3% of primary DLBCL), probably originated as by-products of the somatic hypermutation process. No CD95 mutations in the two analyzed regions were detected in primary extranodal DLBCL, mediastinal large B-cell lymphoma (MLBCL), and DLBCL arising from indolent low-grade lymphomas. Because of our results, a review of published data with respect to the site of mutations was performed, which suggested a different distribution of mutations in nodal and extranodal DLBCL.
Collapse
Affiliation(s)
- Vanesa Scholl
- Bone Marrow Transplantation Centre (CEMO), Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | | | | | | | | |
Collapse
|
6
|
Kondo E, Yoshino T. Expression of apoptosis regulators in germinal centers and germinal center-derived B-cell lymphomas: insight into B-cell lymphomagenesis. Pathol Int 2007; 57:391-7. [PMID: 17587238 DOI: 10.1111/j.1440-1827.2007.02115.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Germinal centers (GC) are unique sites in peripheral lymphoid tissue where clonal selection of B cells takes place in response to stimulation by various antigens. To select a proper B-cell clone for antibody-mediated immunity, multiple apoptotic signals synchronize in the GC, both in negative and positive selection pathways. At the same time, GC have been known to be a major source of B-cell lymphomas including follicular and Burkitt's, and also some diffuse large B-cell lymphomas. Therefore, uncovering the biological characteristics of GC would greatly contribute to understanding lymphomagenesis, or progression of B-cell lymphomas of GC origin. Herein the authors briefly explain the expression and pathophysiological significance of apoptosis regulators in GC, focusing particularly on Bcl-2, Fas (CD95) and a transcription factor, nuclear factor of activated T cells, which seems to play a critical role in regulating cellular dynamics of GC B cells via B-cell antigen receptor. The expression of these molecules is then compared with that of the neoplastic counterpart B-cell lymphomas in order to consider lymphomagenesis of GC origin. In conclusion, follicular lymphoma closely reflected characteristics of GC among these B-cell lymphomas, although it acquires strong expression of apoptosis-resistant gene, bcl-2.
Collapse
MESH Headings
- Apoptosis/physiology
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Gene Expression Regulation, Neoplastic
- Germinal Center/metabolism
- Humans
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/metabolism
- Lymphoma, B-Cell/pathology
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/metabolism
- Signal Transduction
- fas Receptor/genetics
- fas Receptor/metabolism
Collapse
Affiliation(s)
- Eisaku Kondo
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | | |
Collapse
|
7
|
Bénéteau M, Daburon S, Moreau JF, Taupin JL, Legembre P. Dominant-negative Fas mutation is reversed by down-expression of c-FLIP. Cancer Res 2007; 67:108-15. [PMID: 17210689 DOI: 10.1158/0008-5472.can-06-1415] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Fas triggering by agonistic antibodies or by its cognate ligand, FasL, induces apoptotic cell death, whereas mutation in the Fas death domain is associated with lymphoma progression. On prolonged culture in the presence of an agonistic anti-Fas antibody, we raised a Jurkat cell line resistant to agonistic antibodies but still sensitive to soluble FasL, which carried at the heterozygous state, a point mutation into the Fas death domain. Down-modulation of c-FLIP expression reversed the blockade of the Fas pathway. We show that the activation threshold for the Fas receptor is more easily overcome by multimeric FasL than by agonistic antibodies and that the increase of this threshold due to mutation in the Fas death domain can be overcome by acting on a downstream effector of the Fas signal, c-FLIP. These findings put forward a new approach to eradicate Fas-resistant tumor cells.
Collapse
Affiliation(s)
- Marie Bénéteau
- Laboratoire Composantes Innées de la Réponse Immunitaire et Différenciation, Centre National de la Recherche Scientifique UMR 5164, University of Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux, France
| | | | | | | | | |
Collapse
|
8
|
Eser B, Sari I, Canoz O, Altuntas F, Cakmak E, Ozturk A, Ozkan M, Er O, Cetin M, Unal A. Prognostic significance of Fas (CD95/APO-1) positivity in patients with primary nodal diffuse large B-cell lymphoma. Am J Hematol 2006; 81:307-314. [PMID: 16628716 DOI: 10.1002/ajh.20564] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Fas (CD95/APO-1) is a protein that is mainly related to apoptosis of lymphoid cells. The increment of Fas expression is associated with long-term survival in various malignancies. However, there are limited studies regarding the effect of Fas expression on the course and prognosis of non-Hodgkin's lymphoma. The aim of this study was to investigate the significance of immunohistochemical Fas expression on the prognosis of nodal diffuse large B-cell lymphoma. A total of 63 patients with primary nodal diffuse large B-cell lymphoma diagnosed in the Erciyes University Department of Hematology between 1990 and 2003 were included in the study. The median age of the patients was 55 years old (range 19-102 years old). The median follow-up period was 19 months (2-132 months). Histopathological sections were stained immunohistochemically and evaluated by light microscopy for Fas, bcl-2, and p53. Clinical and laboratory parameters including Fas, bcl-2, and p53 positivity, age, sex, performance status, clinical stage, presence of B symptoms, bone marrow involvement, extranodal involvement, and lactic dehydrogenase levels were evaluated to compare overall survival. Complete remission was obtained in 28 patients (44.4%) after first-line chemotherapy. Fas positivity, male gender, good performance status, clinical stage I-II, absence of B symptoms, normal lactic dehydrogenase value, and absence of bone marrow involvement were favorable prognostic factors for complete remission in statistical analysis. Multivariate analysis revealed that positive Fas expression and ECOG performance status were independent prognostic factors for overall survival. Also, Fas-positive patients had significantly prolonged progression-free survival. Immunohistochemical Fas positivity was a favorable prognostic factor for complete remission and overall and progression-free survival in primary nodal diffuse large B-cell lymphoma.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Disease-Free Survival
- Female
- Humans
- Immunohistochemistry
- Lymphoma, B-Cell/diagnosis
- Lymphoma, B-Cell/mortality
- Lymphoma, B-Cell/therapy
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/therapy
- Male
- Middle Aged
- Multivariate Analysis
- Neoplasm Staging
- Predictive Value of Tests
- fas Receptor/analysis
- fas Receptor/biosynthesis
Collapse
Affiliation(s)
- Bulent Eser
- Department of Hematology-Oncology, Erciyes University School of Medicine, M.K. Dedeman Oncology Hospital, Kayseri, Turkey.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Hougardy BMT, van der Zee AGJ, van den Heuvel FAJ, Timmer T, de Vries EGE, de Jong S. Sensitivity to Fas-mediated apoptosis in high-risk HPV-positive human cervical cancer cells: relationship with Fas, caspase-8, and Bid. Gynecol Oncol 2005; 97:353-64. [PMID: 15863130 DOI: 10.1016/j.ygyno.2005.01.036] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2004] [Revised: 01/18/2005] [Accepted: 01/31/2005] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Binding of Fas ligand or agonistic anti-Fas antibody to the death receptor Fas can activate a caspase-cascade resulting in apoptosis. In the present study, the functionality of the Fas pathway was studied in human cervical cancer cells with different HPV and p53 status. METHODS HeLa (HPV-18 positive), CaSki, and SiHa (both HPV-16 positive) contain wild-type p53, while C33A (HPV negative) expresses mutant p53. Fas cell surface expression was determined by flow cytometry. Expression of proteins involved in the apoptotic pathway was analyzed by Western blotting and apoptosis was measured by acridine orange staining of nuclear chromatin. RESULTS Despite high Fas membrane expression in the HPV-positive cells, CaSki was highly sensitive, HeLa slightly sensitive, and SiHa and C33A were resistant for agonistic anti-Fas antibody. Almost undetectable Fas membrane levels can explain the non-responsiveness of C33A for anti-Fas. Although interferon-gamma (IFNgamma) strongly and cisplatin to a lesser extend enhanced Fas membrane expression in all HPV-positive cells, sensitization to anti-Fas by IFNgamma or cisplatin was only observed in HeLa. Analysis of the Fas apoptotic pathway showed that anti-Fas treatment induced caspase-8 activation and concomitantly Bid cleavage, caspase-9 and caspase-3 activation, PARP cleavage and apoptosis in HeLa and CaSki. IFNgamma plus anti-Fas treatment, in contrast to anti-Fas alone, facilitated caspase-8 activation in HeLa and SiHa, while an increase in Bid cleavage, caspase-9 activation and apoptosis was only observed in HeLa. Apoptotic failure in SiHa (even in the presence of IFNgamma) was probably due to low caspase-8, almost undetectable Bid protein levels and therefore lack of caspase-9 activation. CONCLUSION Sensitivity to anti-Fas depends on Fas, caspase-8, and Bid protein levels in cervical cancer cells. Additionally, IFNgamma and cisplatin can increase sensitivity to anti-Fas in a subset of HPV-positive cervical cancer cell lines by upregulation of Fas and caspase-8 expression without major changes in p53 levels.
Collapse
Affiliation(s)
- Brigitte M T Hougardy
- Department of Gynecology-Oncology, University Medical Center Groningen, P.O. Box 30001, 9700 RB Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|
10
|
Lundin KU, Screpanti V, Omholt H, Hofgaard PO, Yagita H, Grandien A, Bogen B. CD4+ T cells kill Id+ B-lymphoma cells: FasLigand-Fas interaction is dominant in vitro but is redundant in vivo. Cancer Immunol Immunother 2004; 53:1135-45. [PMID: 15696611 PMCID: PMC11032948 DOI: 10.1007/s00262-004-0538-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2004] [Accepted: 03/19/2004] [Indexed: 01/30/2023]
Abstract
B-lymphoma cells express a highly tumor-specific antigen, monoclonal Ig, which is a promising target for immunotherapy. Previous work has demonstrated that B-lymphoma cells spontaneously process their endogenous monoclonal Ig and present variable (V) region peptides (Id-peptides) on their MHC class II molecules to CD4+ T cells. Id-specific CD4+ T cells protect mice against B-lymphoma cells in the absence of antiidiotypic antibodies. The molecular mechanism by which Id-specific CD4+ T cells kill B-lymphoma cells is hitherto unknown. We here demonstrate in an Id-specific T-cell receptor (TCR)-transgenic mouse model that Id-specific CD4+ T cells induce apoptosis of Fas+ B-lymphoma cells in vitro by FasLigand (FasL)-Fas interaction. Moreover, the rare B lymphomas that had escaped rejection in TCR-transgenic mice had down-regulated their sensitivity to Fas-mediated apoptosis. Although these results suggest that FasL-Fas interaction is important, Id-specific CD4+ T cells could eliminate Id+ B-lymphoma cells in vivo by other mechanisms, since three independent ways of blocking FasL-Fas-mediated killing failed to abrogate tumor protection in TCR-transgenic mice. These results suggest that there are several redundant pathways by which Id-specific CD4+ T cells eliminate Id+ B-lymphoma cells in vivo, of which FasL-Fas interaction is only one.
Collapse
Affiliation(s)
- Katrin U Lundin
- Institute of Immunology, University of Oslo, Rikshospitalet, N-0027 Oslo, Norway.
| | | | | | | | | | | | | |
Collapse
|
11
|
Braess J, Schneiderat P, Schoch C, Fiegl M, Lorenz I, Hiddemann W. Functional analysis of apoptosis induction in acute myeloid leukaemia-relevance of karyotype and clinical treatment response. Br J Haematol 2004; 126:338-47. [PMID: 15257705 DOI: 10.1111/j.1365-2141.2004.05039.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Deficiencies or structural defects of the apoptotic machinery have been postulated as a potential mechanism for a broad resistance of acute myeloid leukaemia (AML) blasts towards cytotoxic therapy comprising chemotherapeutic agents with diverse pharmacodynamic principles but also cell-mediated cytotoxicity of the graft-versus-leukaemia effect, for example, in the setting of allogeneic transplantation. This hypothesis was systematically tested by functionally analysing the early, intermediate and late events of the apoptotic process in primary AML (n = 31) blasts following activation of the intrinsic and extrinsic pathway of apoptosis (etoposide and cytarabine as DNA damaging agents, FAS-ligand as an activator of the death receptor pathway). Activation of the extrinsic pathway by FAS-ligand did not induce apoptosis in primary AML, instead the proapoptotic signal was shown to 'fade', even in the early phase of the apoptotic sequence. However, activation of the intrinsic pathway induced severe cytotoxicity in all samples that showed the characteristic features of typical apoptosis, with a prominent apoptotic volume decrease (blebbing) in the early phase, significant increases in caspase 3 activity (intermediate or effector phase) and breakdown of cellular energy production in the late phase of apoptosis. These characteristics did not differ between prognostically favourable versus unfavourable AML karyotypes or between clinically responding versus refractory AML--indicating that a functional apoptotic apparatus is present even in the unfavourable AML subgroups. Our data indicate that the mechanism for a broad clinical resistance is not a dysfunctional apparatus per se but rather the consequence of anti-apoptotic regulation impeding otherwise functional apoptotic machinery.
Collapse
Affiliation(s)
- Jan Braess
- Department of Internal Medicine III, University Hospital Grosshadern, Ludwig-Maximilians University, Munich, Germany.
| | | | | | | | | | | |
Collapse
|
12
|
Alenzi FQB, Wyse RKH, Altamimi WG. Apoptosis as a tool for therapeutic agents in haematological diseases. Expert Opin Biol Ther 2004; 4:407-20. [PMID: 15006734 DOI: 10.1517/14712598.4.3.407] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Apoptosis, an active mechanism of cell death, is an important process in many biological systems. Apoptosis is thought to contribute to many disease processes. This notion has raised expectations that therapeutic opportunities will naturally follow once a better understanding of these processes has been achieved. The regulation of apoptosis in normal and malignant haematological diseases represents an important therapeutic approach in the treatment of leukaemia and lymphoma. This review summarises recent developments in the clinical manipulation of apoptosis pathways in haematological therapy.
Collapse
Affiliation(s)
- Faris Q B Alenzi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, King Faisal University, PO Box 1982, Dammam 31451, Kingdom of Saudi Arabia.
| | | | | |
Collapse
|
13
|
Abstract
Apoptosis, the cell's intrinsic death program, is a key regulator of tissue homeostasis. An imbalance between cell death and proliferation may result in tumor formation. Also, killing of cancer cells by cytotoxic therapies such as chemotherapy, gamma-irradiation or ligation of death receptors is predominantly mediated by triggering apoptosis in target cells. In addition to the intrinsic mitochondrial pathway, elements of death receptor signaling pathways have been implied to contribute to the efficacy of cancer therapy. Failure to undergo apoptosis in response to anticancer therapy may lead to resistance. Also, deregulated expression of death receptor pathway molecules may contribute to tumorigenesis and tumor escape from endogenous growth control. Understanding the molecular events that regulate apoptosis induced by anticancer therapy and how cancer cells evade apoptosis may provide new opportunities for pathway-based rational therapy and for drug development.
Collapse
|
14
|
Passegué E, Jamieson CHM, Ailles LE, Weissman IL. Normal and leukemic hematopoiesis: are leukemias a stem cell disorder or a reacquisition of stem cell characteristics? Proc Natl Acad Sci U S A 2003; 100 Suppl 1:11842-9. [PMID: 14504387 PMCID: PMC304096 DOI: 10.1073/pnas.2034201100] [Citation(s) in RCA: 445] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Leukemia can be viewed as a newly formed, abnormal hematopoietic tissue initiated by a few leukemic stem cells (LSCs) that undergo an aberrant and poorly regulated process of organogenesis analogous to that of normal hematopoietic stem cells. A hallmark of all cancers is the capacity for unlimited self-renewal, which is also a defining characteristic of normal stem cells. Given this shared attribute, it has been proposed that leukemias may be initiated by transforming events that take place in hematopoietic stem cells. Alternatively, leukemias may also arise from more committed progenitors caused by mutations and/or selective expression of genes that enhance their otherwise limited self-renewal capabilities. Identifying the LSCs for each type of leukemia is a current challenge and a critical step in understanding their respective biologies and may provide key insights into more effective treatments. Moreover, LSC identification and purification will provide a powerful diagnostic, prognostic, and therapeutic tool in the clinic.
Collapse
Affiliation(s)
- Emmanuelle Passegué
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
15
|
Abstract
For the majority of patients with advance malignancies, current therapies are noncurative. Developing therapeutic agents that enhance the apoptotic effects and hence antitumor potential of currently available chemotherapy agents represents a rationale investigative strategy. Several chemotherapeutic agents including antimicrotubule agents and all-trans-retinoic acid utilize these pathways to mediate tumor cell killing. With specific agents such as oblimersan sodium in randomized "pivotal" studies, and agents targeting the TRAIL receptor-family recently entering early clinical study, cautious optimism is warranted.
Collapse
Affiliation(s)
- Anthony W Tolcher
- Institute for Drug Development, Cancer Therapy and Research Center, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
16
|
Bernstorff WV, Glickman JN, Odze RD, Farraye FA, Joo HG, Goedegebuure PS, Eberlein TJ. Fas (CD95/APO-1) and Fas ligand expression in normal pancreas and pancreatic tumors. Implications for immune privilege and immune escape. Cancer 2002; 94:2552-60. [PMID: 12173320 DOI: 10.1002/cncr.10549] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Fas (CD95/APO-1) and Fas ligand (FasL) play key roles in immunologic homeostasis and immune privilege and may regulate normal cell turnover. Earlier studies had suggested that FasL-positive pancreatic carcinoma cell lines can induce apoptosis in T cells, thereby evading host immune surveillance. In the current study the authors have characterized the expression of Fas and FasL in the normal pancreas and in pancreatic neoplasia. METHODS Pancreatic resection specimens with ductal-type adenocarcinoma or intraductal dysplasia (n = 41), nonductal pancreatic neoplasms (n = 5), and chronic pancreatitis (n = 4) were examined for Fas and FasL expression by immunohistochemistry. The results in invasive adenocarcinoma were compared to those for benign ducts and intraductal dysplasia, and correlated with clinicopathologic features of the tumors and with patient survival. RESULTS Fas was expressed in the normal pancreatic ducts and in intraductal dysplasia in a mixed membrane/cytoplasmic pattern. In all cases of invasive ductal-type adenocarcinoma, membranous Fas could not be detected; cytoplasmic Fas staining was reduced or completely lost. Loss of Fas expression in pancreatic ductal-type adenocarcinomas significantly correlated with poorer differentiation and extrapancreatic spread of the tumors and was associated with a shorter overall survival. FasL expression was present in the normal pancreatic ducts as well as in islet cells and was maintained in all pancreatic tumors. CONCLUSIONS These results implicate the Fas pathway in the regulation of physiologic cell turnover and immune privilege in the normal pancreas and indicate that loss of Fas expression is correlated with malignant transformation and biologic aggressiveness in pancreatic adenocarcinomas. This may represent a mechanism by which pancreatic tumor cells become resistant to apoptosis and escape immune surveillance in vivo.
Collapse
Affiliation(s)
- Wolfram V Bernstorff
- Laboratory of Biologic Cancer Therapy, Division of Surgical Oncology, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Apoptosis plays a critical role in lymphocyte development and homeostasis. Enhanced lymphocyte apoptosis can cause immunodeficiency through cell loss. Conversely, inhibition of apoptosis can lead to the development of autoimmunity or lymphoma. Two major pathways contribute to the regulation of lymphocyte cell death, death-by-neglect and death-by-instruction.
Collapse
Affiliation(s)
- Jeffrey C Rathmell
- Abramson Family Cancer Research Institute, Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
18
|
Shain KH, Landowski TH, Dalton WS. Adhesion-mediated intracellular redistribution of c-Fas-associated death domain-like IL-1-converting enzyme-like inhibitory protein-long confers resistance to CD95-induced apoptosis in hematopoietic cancer cell lines. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:2544-53. [PMID: 11859150 DOI: 10.4049/jimmunol.168.5.2544] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Evasion of immune surveillance is a key step in malignant progression. Interactions between transformed hematopoietic cells and their environment may initiate events that confer resistance to apoptosis and facilitate immune evasion. In this report, we demonstrate that beta(1) integrin-mediated adhesion to fibronectin inhibits CD95-induced caspase-8 activation and apoptosis in hematologic tumor cell lines. This adhesion-dependent inhibition of CD95-mediated apoptosis correlated with enhanced c-Fas-associated death domain-like IL-1-converting enzyme-like inhibitory protein-long (c-FLIP(L)) cytosolic solubility compared with nonadhered cells. Cytosolic c-FLIP(L) protein preferentially associated with cytosolic Fas-associated death domain protein (FADD) and localized to the death-inducing signal complex after CD95 ligation in adherent cells. The incorporation of c-FLIP(L) in the death-inducing signal complex prevented procaspase-8 processing and activation of the effector phase of apoptosis. Adhesion to fibronectin increased c-FLIP(L) cytosolic solubility and availability for FADD binding by redistributing c-FLIP(L) from a preexisting membrane-associated fraction. Increased cytosolic availability of c-FLIP(L) for FADD binding was not related to increased levels of RNA or protein synthesis. These data show that adhesion of anchorage-independent cells to fibronectin provides a novel mechanism of resistance to CD95-mediated programmed cell death by regulating the cellular localization and availability of c-FLIP(L).
Collapse
Affiliation(s)
- Kenneth H Shain
- Department of Interdisciplinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL 33612, USA
| | | | | |
Collapse
|