Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Obes Surg ; 29(4): 1309-1316, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30689172

RESUMO

BACKGROUND: Novice surgeons experience high levels of physical and mental workload during the early stages of their curriculum and clinical practice. Laparoscopic sleeve gastrectomy is the first bariatric procedure worldwide. Feasibility and safety of single-port sleeve gastrectomy (SPSG) has been demonstrated. An immersive virtual reality (VR) simulation was developed to provide a repetitive exercise to learn this novel technique. The primary objective of this study was to evaluate the impact of the VR training tool on mental and physical workload in novice surgeons. The secondary objective included an evaluation of the VR simulator. METHODS: A monocentric-controlled trial was conducted. Ten participants were divided into two groups, the VR group and the control group (without VR training). Surgery residents participated in a first real case of SPSG and a second case 1 month later. The VR group underwent a VR training between the two surgeries. Mental and physical loads were assessed with self-assessment questionnaires: NASA-TLX, Borg scale, and manikin discomfort test. The VR simulator was evaluated through presence, cybersickness, and usability questionnaires. RESULTS: This study showed a decrease of the mental demand and effort dimensions of NASA-TLX between the first and the second surgery in the VR group (P < .05). During the second surgery, a marginally significant difference was shown concerning the mental demand between the two groups. Postural discomfort of the VR group decreased with practice (P < .01), mainly between the first and the second surgery (P < .05). Furthermore, participants characterized the VR simulator as realistic, usable, and very useful to learned surgery. CONCLUSION: This exploratory study showed an improvement in mental and physical workload when novice surgeons trained with VR (repetitive practice, gesture improvement, reduction of stress, etc.). Virtual reality appears to be a promising perspective for surgical training.


Assuntos
Gastrectomia/educação , Internato e Residência , Obesidade Mórbida/cirurgia , Cirurgiões/educação , Realidade Virtual , Carga de Trabalho , Adulto , Esgotamento Profissional/etiologia , Esgotamento Profissional/prevenção & controle , Esgotamento Psicológico/etiologia , Esgotamento Psicológico/prevenção & controle , Competência Clínica , Currículo , Feminino , Gastrectomia/métodos , Humanos , Satisfação no Emprego , Laparoscopia/educação , Laparoscopia/métodos , Masculino , Autoavaliação (Psicologia) , Treinamento por Simulação/métodos , Inquéritos e Questionários , Carga de Trabalho/psicologia
2.
Oncoimmunology ; 7(8): e1463947, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30221060

RESUMO

The immune system avoids oncogenesis and slows down tumor progression through a mechanism called immunosurveillance. Nevertheless, some malignant cells manage to escape from immune control and form clinically detectable tumors. Tetraploidy, which consists in the intrinsically unstable duplication of the genome, is considered as a (pre)-cancerous event that can result in aneuploidy and contribute to oncogenesis. We previously described the fact that tetraploid cells can be eliminated by the immune system. Here, we investigate the role of different innate and acquired immune effectors by inoculating hyperploid cancer cells into wild type or mice bearing different immunodeficient genotypes (Cd1d-/-, FcRn-/-, Flt3l-/-, Foxn1nu/nu, MyD88-/-, Nlrp3-/-, Ighmtm1Cgn, Rag2-/-), followed by the monitoring of tumor incidence, growth and final ploidy status. Our results suggest that multiple different immune effectors including B, NK, NKT and T cells, as well as innate immune responses involving the interleukine-1 receptor and the Toll-like receptor systems participate to the immunoselection against hyperploid cells. Hence, optimal anticancer immunosurveillance likely involves the contribution of multiple arms of the immune system.

3.
Oncoimmunology ; 1(3): 271-278, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22737602

RESUMO

The success of anticancer chemotherapy relies at least in part on the induction of an immune response against tumor cells. Thus, tumors growing on mice that lack the pattern recognition receptor TLR4 or the purinergic receptor P2RX7 fail to respond to chemotherapy with anthracyclins or oxaliplatin in conditions in which the same neoplasms growing on immunocompetent mice would do so. Similarly, the therapeutic efficacy (measured as progression-free survival) of adjuvant chemotherapy with anthracyclins is reduced in breast cancer patients bearing loss-of-function alleles of TLR4 or P2RX7. TLR4 loss-of-function alleles also have a negative impact on the therapeutic outcome of oxaliplatin in colorectal cancer patients. Here, we report that loss-of-function TLR4 and P2RX7 alleles do not affect overall survival in non-small cell lung cancer (NSCLC) patients, irrespective of the administration and type of chemotherapy. The intrinsic characteristics of NSCLC (which near-to-always is chemoresistant and associated with poor prognosis) and/or the type of therapy that is employed to treat this malignancy (which near-to-always is based on cisplatin) may explain why two genes that affect the immune response to dying cells fail to influence the clinical progression of NSCLC patients.

4.
Eur J Endocrinol ; 165(3): 365-73, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21646289

RESUMO

Adrenalectomy for pheochromocytoma is reported with a mortality close to zero in recent studies. The dogma of preoperative fluid and hypotensive drug administrations is widely applied in patients scheduled for pheochromocytoma removal and is assumed to have a beneficial effect on operative outcomes. This paradigm is only based on historical studies of non-standardized practices and criteria for efficacy, with no control group. Pre- and intraoperative hypovolemia have never been demonstrated in patients scheduled for pheochromocytoma removal. Recent improvements in outcome of patients undergoing adrenalectomy for pheochromocytoma could also be the result of improvement in surgical techniques and refinement in anesthetic practices. Whether better knowledge of the disease, efficiency of available intravenous short-acting vasoactive drugs, and careful intraoperative handling of the tumor make it possible to omit preoperative preparation in most patients scheduled for pheochromocytoma removal is presently questionable. We reviewed available literature in this respect.


Assuntos
Neoplasias das Glândulas Suprarrenais/cirurgia , Assistência Perioperatória , Feocromocitoma/cirurgia , Adrenalectomia/efeitos adversos , Feminino , Humanos , Hipertensão/prevenção & controle , Cuidados Intraoperatórios , Gravidez , Complicações Neoplásicas na Gravidez/cirurgia
5.
J Exp Med ; 208(3): 491-503, 2011 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-21383056

RESUMO

By triggering immunogenic cell death, some anticancer compounds, including anthracyclines and oxaliplatin, elicit tumor-specific, interferon-γ-producing CD8(+) αß T lymphocytes (Tc1 CTLs) that are pivotal for an optimal therapeutic outcome. Here, we demonstrate that chemotherapy induces a rapid and prominent invasion of interleukin (IL)-17-producing γδ (Vγ4(+) and Vγ6(+)) T lymphocytes (γδ T17 cells) that precedes the accumulation of Tc1 CTLs within the tumor bed. In T cell receptor δ(-/-) or Vγ4/6(-/-) mice, the therapeutic efficacy of chemotherapy was compromised, no IL-17 was produced by tumor-infiltrating T cells, and Tc1 CTLs failed to invade the tumor after treatment. Although γδ T17 cells could produce both IL-17A and IL-22, the absence of a functional IL-17A-IL-17R pathway significantly reduced tumor-specific T cell responses elicited by tumor cell death, and the efficacy of chemotherapy in four independent transplantable tumor models. Adoptive transfer of γδ T cells restored the efficacy of chemotherapy in IL-17A(-/-) hosts. The anticancer effect of infused γδ T cells was lost when they lacked either IL-1R1 or IL-17A. Conventional helper CD4(+) αß T cells failed to produce IL-17 after chemotherapy. We conclude that γδ T17 cells play a decisive role in chemotherapy-induced anticancer immune responses.


Assuntos
Antineoplásicos/farmacologia , Interleucina-17/fisiologia , Receptores de Antígenos de Linfócitos T gama-delta/fisiologia , Sarcoma Experimental/imunologia , Subpopulações de Linfócitos T/fisiologia , Animais , Antineoplásicos/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/fisiologia , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Morte Celular/fisiologia , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Interferon gama/imunologia , Interferon gama/fisiologia , Interleucina-17/imunologia , Interleucina-23/imunologia , Interleucina-23/fisiologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/fisiopatologia , Transdução de Sinais/imunologia , Transdução de Sinais/fisiologia , Subpopulações de Linfócitos T/imunologia , Resultado do Tratamento
6.
Ann N Y Acad Sci ; 1209: 99-108, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20958322

RESUMO

The concept of immunogenic chemotherapy that has recently emerged relies upon the capacity of a cytotoxic compound to trigger a cell-death modality. This modality elicits cross-priming by dendritic cells of tumor antigen-specific T cells that will contribute to the tumoricidal activity of the compound and protect the host against relapse. In contrast, most anticancer drugs elicit nonimmunogenic apoptosis that is not accompanied with an immunizing property. This review will discuss some molecular and metabolic changes required at the level of the tumor that must engage key pathways at the level of the host for the induction of Tc1 polarized-protective T cell responses during chemotherapy. We will summarize the immune adjuvants that can boost the immunogenicity of cell death to augment the efficacy of chemotherapy.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Animais , Vacinas Anticâncer/administração & dosagem , Terapia Combinada , Humanos , Camundongos , Neoplasias/imunologia , Neoplasias/patologia
7.
Cell Cycle ; 9(15): 3072-7, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20699648

RESUMO

In response to immunogenic cell death inducers, calreticulin (CRT) translocates from its orthotopic localization in the lumen of the endoplasmic reticulum (ER) to the surface of the plasma membrane where it serves as an engulfment signal for antigen-presenting cells.(1) Here, we report that yet another ER protein, the lysyl-tRNA synthetase (KARS), was exposed on the surface of stressed cells, on which KARS co-localized with CRT in lipid rafts. Depletion of KARS with small interfering RNAs suppressed CRT exposure induced by anthracyclines or UVC light. In contrast to CRT, KARS was also found in the supernatant of stressed cells. Recombinant KARS protein was unable to influence the binding of recombinant CRT to the cell surface. Moreover, recombinant KARS protein was unable to stimulate macrophages in vitro. These results underscore the contribution of KARS to the emission of (one of) the principal signal(s) of immunogenic cell death, CRT exposure.


Assuntos
Calreticulina/metabolismo , Membrana Celular/metabolismo , Citotoxicidade Imunológica , Lisina-tRNA Ligase/metabolismo , Sequência de Aminoácidos , Antraciclinas/farmacologia , Morte Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Citotoxicidade Imunológica/efeitos dos fármacos , Células HeLa , Humanos , Lisina-tRNA Ligase/química , Dados de Sequência Molecular , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
8.
Cell Cycle ; 9(14): 2830-5, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20647758

RESUMO

The generation of specific T lymphocyte subsets is under the strict control of specific transcription factors, as this has been shown by knockout experiments in mice. Here, we show that siRNAs that specifically target the transcription factor Gata3 (which is required for the development of T helper 1 cells) or T-Bet (which is required for the development of T helper 2 cells) can be effective in vivo. Thus, the intraperitoneal injection of siRNAs specific for Gata3 or t-Bet leads to the specific depletion of their target gene products in vivo, in the spleen and in the lymph nodes of mice. The immunomodulatory action of these siRNAs was validated in a model of anti-tumor vaccination in which colorectal cancer cells that succumb to anthracyclin-induced immunogenic cell death were injected subcutaneously into one flank, in the absence of any adjuvant and live tumor cells were injected simultaneously in the opposite flank of immunocompetent mice. In this setting, the siRNA targeting t-Bet was able to accelerate tumor growth while the siRNA targeting Gata3 significantly reduced the proliferation of cancer cells in vivo. These effects were dependent on the immune response elicited by dying tumor cells because both siRNAs failed to modulate the growth of tumors in non-vaccinated mice. The immune response-dependent anticancer effect of the Gata3-specific siRNA was not due to the induction of class I interferons and could be fully abolished by co-injection of t-Bet-specific siRNA. These results demonstrate the possibility to use siRNAs for immunomodulaton in vivo and illustrate the antagonistic implication of distinct T helper populations in anti-cancer immune responses.


Assuntos
Fator de Transcrição GATA3/antagonistas & inibidores , Interferência de RNA , Linfócitos T/imunologia , Animais , Linhagem Celular Tumoral , Fator de Transcrição GATA3/genética , Fator de Transcrição GATA3/metabolismo , Camundongos , RNA Interferente Pequeno/metabolismo , Células Th2/imunologia , Células Th2/metabolismo
9.
Semin Immunol ; 22(3): 113-24, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20403709

RESUMO

An attractive, yet hitherto unproven concept predicts that the promotion of tumor regression should elicit the host's immune response against residual tumor cells to achieve an optimal therapeutic effect. In a way, chemo- or radiotherapy must trigger "danger signals" emitted from immunogenic cell death and hence elicit "danger associated molecular patterns" to stimulate powerful anticancer immune responses. Here, based on the recent experimental and clinical evidence, we will discuss the molecular identity of the multiple checkpoints that dictate the success of "immunogenic chemotherapy" at the levels of the drug, of the tumor cell and of the host immune system.


Assuntos
Tratamento Farmacológico , Imunoterapia/métodos , Neoplasias , Radioterapia , Vacinas Anticâncer/imunologia , Humanos , Sistema Imunitário , Interferon gama/metabolismo , Interleucina-1beta/metabolismo , Neoplasias/imunologia , Neoplasias/terapia
10.
Cancer Res ; 70(3): 855-8, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20086177

RESUMO

By destroying tumor cells, conventional anticancer therapies may stimulate the host immune system to eliminate residual disease. Anthracyclines, oxaliplatin, and ionizing irradiation activate a type of tumor cell death that elicits efficient anticancer immune responses depending on interferon gamma (IFNgamma) and the IFNgamma receptor. Thus, dying tumor cells emit danger signals that are perceived by dendritic cells (DC), which link innate and cognate immune responses. Recently, we observed that ATP was released by tumor cells succumbing to chemotherapy. ATP activates purinergic P2RX7 receptors on DC, thus activating the NLRP3/ASC/caspase-1 inflammasome and driving the secretion of interleukin-1beta (IL-1beta). IL-1beta then is required for the adequate polarization of IFNgamma-producing CD8(+) T cells. These results imply a novel danger signal, ATP, and a novel receptor, P2RX7, in the chemotherapy-elicited anticancer immune response.


Assuntos
Trifosfato de Adenosina/metabolismo , Células Dendríticas/imunologia , Imunidade/imunologia , Neoplasias/imunologia , Animais , Morte Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Humanos , Interferon gama/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Receptores de Interferon/metabolismo , Transdução de Sinais/imunologia , Receptor de Interferon gama
11.
Cell Cycle ; 8(23): 3971-7, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19901557

RESUMO

In response to some chemotherapeutic agents, tumor cells can translocate calreticulin (CRT), which is usually contained in the lumen of the endoplasmic reticulum, to the surface of the plasma membrane. This effect requires the phosphorylation of the eukaryotic initiation factor 2alpha(eIF2alpha) by the eIF2alpha kinase PERK, yet may also be triggered by inhibition of the eIF2alpha phosphatase, which is composed by a catalytic subunit (PP1) and a regulatory subunit (GADD34). Here, we addressed the question whether the dissociation of the PP1/GADD34 complex would be sufficient to trigger CRT exposure. Molecular modeling led to the design of a GADD34-derived peptide that competitively disrupts the PP1/GADD34 complex. When added to intact cells, the GADD34-derived peptide fused to a plasma membrane translocation domain abolished the interaction between PP1 and GADD34, stimulated the phosphorylation of eIF2alpha, and triggered CRT exposure. However, the resolution of the PP1/GADD34 complex did not evoke apoptosis, allowing for the dissociation of CRT exposure and cell death. Anthracyclins, which are highly efficient in inducing CRT translocation to the cell surface also stimulated the dissociation of the PP1/GADD34 complex. These results suggest that the PP1/GADD34 complex plays a major role in the regulation of CRT exposure.


Assuntos
Antígenos de Diferenciação/metabolismo , Calreticulina/fisiologia , Proteínas de Ciclo Celular/metabolismo , Proteína Fosfatase 1/metabolismo , Sequência de Aminoácidos , Antraciclinas/farmacologia , Apoptose , Sítios de Ligação , Calreticulina/metabolismo , Simulação por Computador , Retículo Endoplasmático/fisiologia , Fator de Iniciação 2 em Eucariotos/metabolismo , Células HeLa , Humanos , Dados de Sequência Molecular , Fosforilação , eIF-2 Quinase/metabolismo
12.
Cell Cycle ; 8(22): 3723-8, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19855167

RESUMO

Chemotherapy can induce anticancer immune responses. In contrast to a widely extended prejudice, apoptotic cell death is often more efficient in eliciting a protective anticancer immune response than necrotic cell death. Recently, we have found that purinergic receptors of the P2X7 type are required for the anticancer immune response induced by chemotherapy. ATP is the endogenous ligand that has the highest affinity for P2X7. Therefore, we investigated the capacity of a panel of chemotherapeutic agents to induce ATP release from cancer cells. Here, we describe that multiple distinct anticancer drugs reduce the intracellular concentration of ATP before and during the manifestation of apoptotic characteristics such as the dissipation of the mitochondrial transmembrane potential and the exposure of phosphatidylserine residues on the plasma membrane. Indeed, as apoptosis progresses, intracellular ATP concentrations decrease, although even advanced-stage apoptotic cells still contain sizeable ATP levels. Only when cells enter secondary necrosis, the ATP concentration falls to undetectable levels. Concomitantly, a wide range of chemotherapeutic agents causes the release of ATP into the extracellular space as they induce tumor cell death. Hence, ATP release is a general correlate of apoptotic cell death induced by conventional anticancer therapies.


Assuntos
Trifosfato de Adenosina/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Animais , Cádmio/farmacologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Etoposídeo/farmacologia , Citometria de Fluxo , Imunofluorescência , Camundongos , Mitomicina/farmacologia , Mitoxantrona/farmacologia , Compostos Organoplatínicos/farmacologia , Oxaliplatina , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Tapsigargina/farmacologia
13.
Ann N Y Acad Sci ; 1174: 51-60, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19769736

RESUMO

Conventional cancer treatments mediate their effects via the direct elimination of tumor cells. Nonetheless, recent evidence indicates that radiotherapy and some chemotherapeutic agents can also induce specific immune responses that contribute to therapeutic outcomes. Two major tumor-intrinsic changes that determine the immune response against tumors have been identified: the translocation of calreticulin to the plasma membrane and the release of high-mobility group box 1 protein. Together, these changes improve engulfment and processing of apoptotic bodies by dendritic cells, which are involved in the cross-priming of antitumor T lymphocytes in vivo. We review these two molecular mechanisms that dictate the radio/chemotherapy-elicited antitumor immune response and discuss how this knowledge can be clinically exploited to predict and also ameliorate the success of chemo/radiotherapy.


Assuntos
Células Dendríticas/imunologia , Neoplasias/imunologia , Animais , Antineoplásicos/uso terapêutico , Apoptose , Morte Celular/imunologia , Terapia Combinada , Modelos Animais de Doenças , Retículo Endoplasmático/imunologia , Proteína HMGB1/imunologia , Humanos , Imunoterapia/métodos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias/patologia , Neoplasias/terapia , Fagocitose , Isomerases de Dissulfetos de Proteínas/imunologia , Linfócitos T/imunologia
14.
Nat Med ; 15(10): 1170-8, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19767732

RESUMO

The therapeutic efficacy of anticancer chemotherapies may depend on dendritic cells (DCs), which present antigens from dying cancer cells to prime tumor-specific interferon-gamma (IFN-gamma)-producing T lymphocytes. Here we show that dying tumor cells release ATP, which then acts on P2X(7) purinergic receptors from DCs and triggers the NOD-like receptor family, pyrin domain containing-3 protein (NLRP3)-dependent caspase-1 activation complex ('inflammasome'), allowing for the secretion of interleukin-1beta (IL-1beta). The priming of IFN-gamma-producing CD8+ T cells by dying tumor cells fails in the absence of a functional IL-1 receptor 1 and in Nlpr3-deficient (Nlrp3(-/-)) or caspase-1-deficient (Casp-1(-/-)) mice unless exogenous IL-1beta is provided. Accordingly, anticancer chemotherapy turned out to be inefficient against tumors established in purinergic receptor P2rx7(-/-) or Nlrp3(-/-) or Casp1(-/-) hosts. Anthracycline-treated individuals with breast cancer carrying a loss-of-function allele of P2RX7 developed metastatic disease more rapidly than individuals bearing the normal allele. These results indicate that the NLRP3 inflammasome links the innate and adaptive immune responses against dying tumor cells.


Assuntos
Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Interleucina-1beta/metabolismo , Animais , Caspase 1/metabolismo , Células Dendríticas/citologia , Imunidade/imunologia , Imunidade Inata , Inflamação/imunologia , Camundongos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Timoma/imunologia
15.
Cell Cycle ; 8(6): 860-9, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19221507

RESUMO

While physiological cell death is non-immunogenic, pathogen induced cell death can be immunogenic and hence stimulate an immune response against antigens that derive from dying cells and are presented by dendritic cells (DCs). The obligate immunogenic "eat-me" signal generated by dying cells consists in the exposure of calreticulin (CRT) at the cell surface. This particular "eat-me" signal, which facilitates engulfment by DCs, can only be found on cells that succumb to immunogenic apoptosis, while it is not present on cells dying in an immunologically silent fashion. CRT normally resides in the lumen of the endoplasmic reticulum (ER), yet can translocate to the plasma membrane surface through a complex pathway that involves elements of the ER stress response (e.g., the eIF2alpha-phosphorylating kinase PERK), the apoptotic machinery (e.g., caspase-8 and its substrate BAP31, Bax, Bak), the anterograde transport from the ER to the Golgi apparatus, and SNARE-dependent exocytosis. A large panoply of viruses encodes proteins that inhibit eIF2alpha kinases, catalyze the dephosphorylation of eIF2alpha, bind to caspase-8, Bap31, Bax or Bak, or perturb exocytosis. We therefore postulate that obligate intracellular pathogens have developed a variety of strategies to subvert CRT exposure, thereby avoiding immunogenic cell death.


Assuntos
Apoptose/imunologia , Calreticulina/imunologia , Células Dendríticas/imunologia , Retículo Endoplasmático/imunologia , Vírus/metabolismo , Animais , Humanos , Neoplasias/imunologia , Vírus/genética
16.
Apoptosis ; 14(4): 364-75, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19145485

RESUMO

It is still enigmatic under which circumstances cellular demise induces an immune response or rather remains immunologically silent. Moreover, the question remains open under which circumstances apoptotic, autophagic or necrotic cells are immunogenic or tolerogenic. Although apoptosis appears to be morphologically homogenous, recent evidence suggests that the pre-apoptotic surface-exposure of calreticulin may dictate the immune response to tumor cells that succumb to anticancer treatments. Moreover, the release of high-mobility group box 1 (HMGB1) during late apoptosis and secondary necrosis contributes to efficient antigen presentation and cytotoxic T-cell activation because HMGB1 can bind to Toll like receptor 4 on dendritic cells, thereby stimulating optimal antigen processing. Cell death accompanied by autophagy also may facilitate cross priming events. Apoptosis, necrosis and autophagy are closely intertwined processes. Often, cells manifest autophagy before they undergo apoptosis or necrosis, and apoptosis is generally followed by secondary necrosis. Whereas apoptosis and necrosis irreversibly lead to cell death, autophagy can clear cells from stress factors and thus facilitate cellular survival. We surmise that the response to cellular stress like chemotherapy or ionizing irradiation, dictates the immunological response to dying cells and that this immune response in turn determines the clinical outcome of anticancer therapies. The purpose of this review is to summarize recent insights into the immunogenicity of dying tumor cells as a function of the cell death modality.


Assuntos
Apoptose/imunologia , Calreticulina/imunologia , Imunidade/imunologia , Neoplasias/imunologia , Animais , Autofagia , Morte Celular/imunologia , Humanos , Modelos Biológicos , Necrose , Neoplasias/patologia
17.
Curr Opin Oncol ; 21(1): 71-6, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19125021

RESUMO

PURPOSE OF REVIEW: It is an ongoing conundrum under which circumstances cellular demise induces an immune response and whether apoptotic or necrotic cells are intrinsically immunogenic or tolerogenic. This review summarizes recent insights in the immunogenicity of dying tumor cells. RECENT FINDINGS: Although apoptosis appears to be morphologically homogeneous, recent evidence suggests that the preapoptotic surface exposure of calreticulin (CRT) may have a profound impact on the immune response. Moreover, the release of high mobility group box 1 protein (HMGB1) during late apoptosis promotes antigen processing by dendritic cells and hence contributes to efficient antigen presentation and cytotoxic T-cell activation. HMGB1 is sensed by TLR4 on dendritic cells, and loss-of-function alleles of TLR4 abolish anticancer immune response and accelerate tumor progression. SUMMARY: A combination of signals elicits an efficient immune response against tumor cells that are dying in response to anthracyclines or ionizing irradiation. During early apoptosis, caspase activation and endoplasmic reticulum stress facilitate the surface exposure of immunogenic effectors followed by the release of soluble factors that are indispensable for effective immune response. Failure of tumor cells to expose/secrete such immunogenic factors and/or failure of the immune system to sense such effectors may compromise the efficacy of conventional anticancer therapies.


Assuntos
Apoptose/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Animais , Células Dendríticas/imunologia , Humanos , Transdução de Sinais/imunologia
18.
EMBO J ; 28(5): 578-90, 2009 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-19165151

RESUMO

Dying tumour cells can elicit a potent anticancer immune response by exposing the calreticulin (CRT)/ERp57 complex on the cell surface before the cells manifest any signs of apoptosis. Here, we enumerate elements of the pathway that mediates pre-apoptotic CRT/ERp57 exposure in response to several immunogenic anticancer agents. Early activation of the endoplasmic reticulum (ER)-sessile kinase PERK leads to phosphorylation of the translation initiation factor eIF2alpha, followed by partial activation of caspase-8 (but not caspase-3), caspase-8-mediated cleavage of the ER protein BAP31 and conformational activation of Bax and Bak. Finally, a pool of CRT that has transited the Golgi apparatus is secreted by SNARE-dependent exocytosis. Knock-in mutation of eIF2alpha (to make it non-phosphorylatable) or BAP31 (to render it uncleavable), depletion of PERK, caspase-8, BAP31, Bax, Bak or SNAREs abolished CRT/ERp57 exposure induced by anthracyclines, oxaliplatin and ultraviolet C light. Depletion of PERK, caspase-8 or SNAREs had no effect on cell death induced by anthracyclines, yet abolished the immunogenicity of cell death, which could be restored by absorbing recombinant CRT to the cell surface.


Assuntos
Antineoplásicos/farmacologia , Calreticulina/fisiologia , Morte Celular/imunologia , Retículo Endoplasmático/metabolismo , Antraciclinas/imunologia , Antraciclinas/farmacologia , Antineoplásicos/imunologia , Apoptose , Caspase 3/metabolismo , Caspase 8/metabolismo , Linhagem Celular , Fator de Iniciação 2 em Eucariotos/metabolismo , Exocitose , Complexo de Golgi/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Compostos Organoplatínicos/imunologia , Compostos Organoplatínicos/farmacologia , Oxaliplatina , Fosforilação , Proteínas SNARE/metabolismo , Raios Ultravioleta , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo , eIF-2 Quinase/metabolismo
19.
J Clin Invest ; 118(6): 1991-2001, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18523649

RESUMO

Although the impact of tumor immunology on the clinical management of most cancers is still negligible, there is increasing evidence that anticancer immune responses may contribute to the control of cancer after conventional chemotherapy. Thus, radiotherapy and some chemotherapeutic agents, in particular anthracyclines, can induce specific immune responses that result either in immunogenic cancer cell death or in immunostimulatory side effects. This anticancer immune response then helps to eliminate residual cancer cells (those that fail to be killed by chemotherapy) or maintains micrometastases in a stage of dormancy. Based on these premises, in this Review we address the question, How may it be possible to ameliorate conventional therapies by stimulating the anticancer immune response? Moreover, we discuss the rationale of clinical trials to evaluate and eventually increase the contribution of antitumor immune responses to the therapeutic management of neoplasia.


Assuntos
Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Neoplasias/imunologia , Neoplasias/terapia , Animais , Antineoplásicos/uso terapêutico , Biotecnologia/métodos , Linhagem Celular Tumoral , Ensaios Clínicos como Assunto , Terapia Combinada/métodos , Resistencia a Medicamentos Antineoplásicos , Humanos , Sistema Imunitário , Imunoterapia/métodos , Oncologia/métodos , Camundongos , Modelos Biológicos
20.
Cancer Res ; 68(11): 4026-30, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18519658

RESUMO

The efficacy of anticancer treatments is mostly assessed by their ability to directly inhibit the proliferation of tumor cells. Recently, we showed that tumor cell death triggered by chemotherapy or radiotherapy initiates an immunoadjuvant pathway that contributes to the success of cytotoxic treatments. The interaction of high mobility group box 1 protein (HMGB1) released from dying tumor cells with Toll-like receptor 4 (TLR4) on dendritic cells was required for the crosspresentation of tumor antigens and the promotion of tumor specific cytotoxic T-cell responses. Breast cancer patients harboring the loss-of-function Asp299Gly polymorphism of TLR4 relapsed earlier after receiving anthracycline-based chemotherapy. These data suggests that HMGB1- and TLR4-dependent immune responses elicited by conventional cancer treatment may increase the probability to achieve a durable therapeutic success.


Assuntos
Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Imunidade Inata , Animais , Antraciclinas/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Humanos , Camundongos , Camundongos Nus , Polimorfismo Genético , Ligação Proteica , Radioterapia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA