Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Front Immunol ; 12: 677707, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34017346

RESUMO

Granzymes are a family of serine proteases stored in granules inside cytotoxic cells of the immune system. Granzyme K (GrK) has been only limitedly characterized and knowledge on its molecular functions is emerging. Traditionally GrK is described as a granule-secreted, pro-apoptotic serine protease. However, accumulating evidence is redefining the functions of GrK by the discovery of novel intracellular (e.g. cytotoxicity, inhibition of viral replication) and extracellular roles (e.g. endothelial activation and modulation of a pro-inflammatory immune cytokine response). Moreover, elevated GrK levels are associated with disease, including viral and bacterial infections, airway inflammation and thermal injury. This review aims to summarize and discuss the current knowledge of i) intracellular and extracellular GrK activity, ii) cytotoxic and non-cytotoxic GrK functioning, iii) the role of GrK in disease, and iv) GrK as a potential therapeutic target.


Assuntos
Espaço Extracelular/imunologia , Granzimas/imunologia , Granzimas/metabolismo , Espaço Intracelular/imunologia , Animais , Infecções Bacterianas/tratamento farmacológico , Infecções Bacterianas/imunologia , Infecções Bacterianas/metabolismo , Citocinas/metabolismo , Citotoxicidade Imunológica , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Espaço Extracelular/metabolismo , Granzimas/antagonistas & inibidores , Humanos , Espaço Intracelular/metabolismo , Pneumopatias/tratamento farmacológico , Pneumopatias/imunologia , Pneumopatias/metabolismo , Terapia de Alvo Molecular/métodos , Inibidores de Serina Proteinase/uso terapêutico , Resultado do Tratamento , Viroses/tratamento farmacológico , Viroses/imunologia , Viroses/metabolismo
2.
Theranostics ; 11(8): 3781-3795, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33664861

RESUMO

Aims: Peritonitis is one of the most common causes of sepsis, a serious syndrome characterized by a dysregulated systemic inflammatory response. Recent evidence suggests that Granzyme A (GzmA), a serine protease mainly expressed by NK and T cells, could act as a proinflammatory mediator and could play an important role in the pathogenesis of sepsis. This work aims to analyze the role and the therapeutic potential of GzmA in the pathogenesis of peritoneal sepsis. Methods: The level of extracellular GzmA as well as GzmA activity were analyzed in serum from healthy volunteers and patients with confirmed peritonitis and were correlated with the Sequential Organ Failure Assessment (SOFA) score. Peritonitis was induced in C57Bl/6 (WT) and GzmA-/- mice by cecal ligation and puncture (CLP). Mice were treated intraperitoneally with antibiotics alone or in combination serpinb6b, a specific GzmA inhibitor, for 5 days. Mouse survival was monitored during 14 days, levels of some proinflammatory cytokines were measured in serum and bacterial load and diversity was analyzed in blood and spleen at different times. Results: Clinically, elevated GzmA was observed in serum from patients with abdominal sepsis suggesting that GzmA plays an important role in this pathology. In the CLP model GzmA deficient mice, or WT mice treated with an extracellular GzmA inhibitor, showed increased survival, which correlated with a reduction in proinflammatory markers in both serum and peritoneal lavage fluid. GzmA deficiency did not influence bacterial load in blood and spleen and GzmA did not affect bacterial replication in macrophages in vitro, indicating that GzmA has no role in bacterial control. Analysis of GzmA in lymphoid cells following CLP showed that it was mainly expressed by NK cells. Mechanistically, we found that extracellular active GzmA acts as a proinflammatory mediator in macrophages by inducing the TLR4-dependent expression of IL-6 and TNFα. Conclusions: Our findings implicate GzmA as a key regulator of the inflammatory response during abdominal sepsis and provide solid evidences about its therapeutic potential for the treatment of this severe pathology.


Assuntos
Granzimas/antagonistas & inibidores , Peritonite/tratamento farmacológico , Peritonite/enzimologia , Sepse/tratamento farmacológico , Sepse/enzimologia , Idoso , Idoso de 80 Anos ou mais , Animais , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Granzimas/sangue , Granzimas/deficiência , Granzimas/genética , Humanos , Mediadores da Inflamação/sangue , Interleucina-6/biossíntese , Células Matadoras Naturais/enzimologia , Macrófagos/enzimologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Peritonite/etiologia , Medicina de Precisão , Sepse/etiologia , Serpinas/farmacologia , Receptor 4 Toll-Like/metabolismo
3.
J Invest Dermatol ; 141(3): 575-585, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32738245

RESUMO

In cutaneous leishmaniasis, the immune response is not only protective but also mediates immunopathology. We previously found that cytolytic CD8 T cells promote inflammatory responses that are difficult to treat with conventional therapies that target the parasite. Therefore, we hypothesized that inhibiting CD8 T-cell cytotoxicity would reduce disease severity in patients. IL-15 is a potential target for such a treatment because it is highly expressed in human patients with cutaneous leishmaniasis lesions and promotes granzyme B‒dependent CD8 T-cell cytotoxicity. Here we tested whether tofacitinib, which inhibits IL-15 signaling by blocking Jak3, might decrease CD8-dependent pathology. We found that tofacitinib reduced the expression of granzyme B by CD8 T cells in vitro and in vivo systemic and topical treatment, with tofacitinib protecting mice from developing severe cutaneous leishmaniasis lesions. Importantly, tofacitinib treatment did not alter T helper type 1 responses or parasite control. Collectively, our results suggest that host-directed therapies do not need to be limited to autoimmune disorders and that topical tofacitinib application should be considered a strategy for the treatment of cutaneous leishmaniasis disease in combination with antiparasitic drugs.


Assuntos
Antiparasitários/uso terapêutico , Granzimas/antagonistas & inibidores , Leishmaniose Cutânea/tratamento farmacológico , Piperidinas/uso terapêutico , Pirimidinas/uso terapêutico , Linfócitos T Citotóxicos/efeitos dos fármacos , Transferência Adotiva , Animais , Antiparasitários/farmacologia , Biópsia , Modelos Animais de Doenças , Quimioterapia Combinada/métodos , Granzimas/metabolismo , Humanos , Leishmania braziliensis/efeitos dos fármacos , Leishmania braziliensis/imunologia , Leishmania braziliensis/isolamento & purificação , Leishmaniose Cutânea/diagnóstico , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/parasitologia , Camundongos , Carga Parasitária , Piperidinas/farmacologia , Pirimidinas/farmacologia , Índice de Gravidade de Doença , Pele/efeitos dos fármacos , Pele/imunologia , Pele/parasitologia , Pele/patologia , Linfócitos T Citotóxicos/imunologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia
4.
Cell Rep ; 32(1): 107847, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32640217

RESUMO

If not properly regulated, the inflammatory immune response can promote carcinogenesis, as evident in colorectal cancer (CRC). Aiming to gain mechanistic insight into the link between inflammation and CRC, we perform transcriptomics analysis of human CRC, identifying a strong correlation between expression of the serine protease granzyme A (GzmA) and inflammation. In a dextran sodium sulfate and azoxymethane (DSS/AOM) mouse model, deficiency and pharmacological inhibition of extracellular GzmA both attenuate gut inflammation and prevent CRC development, including the initial steps of cell transformation and epithelial-to-mesenchymal transition. Mechanistically, extracellular GzmA induces NF-κB-dependent IL-6 production in macrophages, which in turn promotes STAT3 activation in cultured CRC cells. Accordingly, colon tissues from DSS/AOM-treated, GzmA-deficient animals present reduced levels of pSTAT3. By identifying GzmA as a proinflammatory protease that promotes CRC development, these findings provide information on mechanisms that link immune cell infiltration to cancer progression and present GzmA as a therapeutic target for CRC.


Assuntos
Carcinogênese/patologia , Colo/patologia , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/patologia , Espaço Extracelular/enzimologia , Granzimas/metabolismo , Inflamação/patologia , Doença Aguda , Animais , Azoximetano , Carcinogênese/genética , Doença Crônica , Neoplasias Colorretais/genética , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , Sulfato de Dextrana , Progressão da Doença , Granzimas/antagonistas & inibidores , Granzimas/genética , Humanos , Inflamassomos/metabolismo , Mediadores da Inflamação/metabolismo , Interleucina-6/biossíntese , Camundongos Knockout , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
5.
Cancer Immunol Res ; 8(4): 506-517, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32075800

RESUMO

The presence and activity of CD8+ T cells within the tumor microenvironment are essential for the control of tumor growth. Utilizing B16-F10 melanoma tumors that express altered peptide ligands of chicken ovalbumin, OVA257-264, we measured high- and low-affinity OVA-specific responses following adoptive transfer of OT-I CD8+ T cell into mice subsequently challenged with tumors. T-cell receptor (TCR) affinity positively correlated with the frequency of OT-I tumor-infiltrating lymphocytes (TIL). Differences in TCR affinity inversely corresponded to in vivo tumor growth rate. Blockade of the PD-1 and CTLA-4 checkpoints preferentially increased the frequency and antitumor function of TIL responding to high-affinity antigens, while failing to enhance the antitumor activity of low-affinity T cells. To determine whether lowering the TCR activation threshold could enhance the breadth and magnitude of the antitumor T-cell response, we inhibited Src homology region 2 domain-containing phosphatase 1 (SHP-1) in OT-I T cells prior to tumor antigen exposure. SHP-1 knockdown increased the cytokine-producing potential of high- and low-affinity T cells but failed to enhance control of tumor growth. In contrast, when SHP-1 knockdown of OT-I T cells was combined with immunotherapy, we observed a significant and long-lasting suppression of tumor growth mediated by low-affinity T cells. We conclude that lowering the TCR activation threshold by targeting SHP-1 expands the repertoire of T cells available to respond to conventional checkpoint blockade, leading to enhanced control of tumor growth.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Granzimas/antagonistas & inibidores , Imunoterapia Adotiva/métodos , Melanoma Experimental/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 6/antagonistas & inibidores , Neoplasias Cutâneas/imunologia , Animais , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Humanos , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/terapia , Microambiente Tumoral
6.
Cancer Immunol Res ; 8(4): 518-529, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32019780

RESUMO

Chimeric antigen receptor (CAR) T-cell therapy is effective in the treatment of cancers of hematopoietic origin. In the immunosuppressive solid tumor environment, CAR T cells encounter obstacles that compromise their efficacy. We developed a strategy to address these barriers by having CAR T cells secrete single-domain antibody fragments [variable heavy domain of heavy chain antibodies (VHH) or nanobodies] that can modify the intratumoral immune landscape and thus support CAR T-cell function in immunocompetent animals. VHHs are small in size and able to avoid domain swapping when multiple nanobodies are expressed simultaneously-features that can endow CAR T cells with desirable properties. The secretion of an anti-CD47 VHH by CAR T cells improves engagement of the innate immune system, enables epitope spreading, and can enhance the antitumor response. CAR T cells that secrete anti-PD-L1 or anti-CTLA-4 nanobodies show improved persistence and demonstrate the versatility of this approach. Furthermore, local delivery of secreted anti-CD47 VHH-Fc fusions by CAR T cells at the tumor site limits their systemic toxicity. CAR T cells can be further engineered to simultaneously secrete multiple modalities, allowing for even greater tailoring of the antitumor immune response.


Assuntos
Antígeno CD47/antagonistas & inibidores , Linfócitos T CD8-Positivos/imunologia , Imunoterapia Adotiva/métodos , Melanoma Experimental/imunologia , Receptores de Antígenos Quiméricos/imunologia , Proteínas Recombinantes/genética , Anticorpos de Domínio Único/imunologia , Animais , Linhagem Celular Tumoral , Granzimas/antagonistas & inibidores , Melanoma Experimental/genética , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptores de Antígenos Quiméricos/genética , Proteínas Recombinantes/metabolismo , Anticorpos de Domínio Único/genética , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/metabolismo , Microambiente Tumoral
7.
Immunol Lett ; 217: 7-14, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31669380

RESUMO

In addition to expressing inhibitory cytokines and suppressive molecules, Treg cells could downplay inflammation by releasing cytotoxic molecules and eliminating proinflammatory immune cells. Colorectal cancer (CRC) is a common malignancy that has led to many cancer-related deaths. In this study, we investigated the cytotoxic aspect of Treg cells in CRC patients. Data showed that tumor-infiltrating FOXP3+ Treg cells expressed granzyme B immediately following resection, indicating that granzyme B-expressing Treg cells were present directly ex vivo. In the tumor-associated lymph nodes (LNs) and circulating lymphocytes, however, granzyme B-expressing Treg cells were only scarcely found. We then attempted to stimulate granzyme B expression in circulating Treg cells. Granzyme B upregulation in Treg cells could not be activated by standard T cell receptor (TCR) activation through anti-CD3/CD28 and IL-2 but required stimulation with bacterial products, such as with heat-killed Staphylococcus aureus. Interestingly, granzyme B expression was highly concentrated in TIM-3+ Treg cells, a Treg subset previously shown to be enriched in the tumor microenvironment and presented increased suppressive capacity. These TIM-3+ Treg cells presented higher cytolytic capacity toward autologous T conventional cells than the TIM-3- Treg cells, in a manner that was dependent on granzyme B but not TIM-3. Overall, we found that granzyme B-expressing Treg cells were enriched in the tumors from CRC patients and had the potential to eliminate autologous T conventional cells.


Assuntos
Neoplasias Colorretais/imunologia , Neoplasias Colorretais/metabolismo , Granzimas/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T/imunologia , Antígenos CD28/metabolismo , Complexo CD3/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/genética , Citocinas/metabolismo , Citotoxicidade Imunológica , Fatores de Transcrição Forkhead/metabolismo , Granzimas/antagonistas & inibidores , Granzimas/genética , Granzimas/imunologia , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Humanos , Interleucina-2/metabolismo , Linfonodos/citologia , Linfonodos/imunologia , Linfonodos/metabolismo , Perforina/metabolismo , Staphylococcus aureus/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo , Microambiente Tumoral/imunologia
8.
Mol Med Rep ; 18(1): 580-586, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29749522

RESUMO

Granzyme B (GrB) is a serine protease that is expressed in the lytic granules of natural killer (NK) cells and cytotoxic T lymphocytes (CTL), and which has been widely reported to serve a crucial role for target cell apoptosis. GrB may serve a non­cytotoxic role in inflammation, but the evidence remains unclear. The present study aimed to establish an inflammatory cell model by using NK92 cells stimulated with lipopolysaccharide (LPS) to investigate whether GrB was involved in the development of inflammation. The extracellular levels of tumor necrosis factor­α (TNF­α), interleukin­1ß (IL­1ß) and GrB were examined by ELISA, and it was demonstrated that LPS treatment increased the extracellular levels of TNF­α, IL­1ß and GrB, and these increased expression levels were inhibited by pretreatment with the GrB inhibitor serpin A3N (SA3N). The protein expression levels of glucose­regulated protein 78 (GRP78), C/EBP homologous protein (CHOP), nuclear factor­κB (NF­κB), inhibitor of NF­κB (IκBα) and GrB were examined by western blot analysis. The results demonstrated that LPS stimulation increased the expression levels of GRP78, CHOP, NF­κB and GrB, and decreased the expression of IκBα, and these changes were inhibited by SA3N, which indicated that inhibition of GrB activity may suppress endoplasmic reticulum (ER) stress signaling. Therefore, it was suggested that GrB may be a potential pro­inflammatory factor, and inhibition of GrB activity may aid the prevention of the development of inflammation by suppressing ER stress signaling.


Assuntos
Estresse do Retículo Endoplasmático , Granzimas/antagonistas & inibidores , Inflamação , Lipopolissacarídeos/farmacologia , Linfócitos/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , Chaperona BiP do Retículo Endoplasmático , Humanos , Lipopolissacarídeos/toxicidade , Linfócitos/efeitos dos fármacos
9.
Trends Cancer ; 3(6): 407-422, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28718416

RESUMO

For more than 20 years perforin and granzymes (GZMs) have been recognized as key cell death executors of cytotoxic T (Tc) and natural killer (NK) cells during cancer immunosurveillance. In immune surveillance, perforin and GZMB, the most potent cytotoxic molecules, act mainly as antitumoral and anti-infectious factors. However, when expressed by immune regulatory cells they may contribute to immune evasion of specific cancer types. By contrast, the other major granzyme, GZMA, seems not to play a major role in Tc/NK cell-mediated cytotoxicity, but acts as a proinflammatory cytokine that might contribute to cancer development. Members of the GZM family also regulate other biological processes unrelated to cell death, such as angiogenesis, vascular integrity, extracellular matrix remodeling, and barrier function, all of which contribute to cancer initiation and progression. Thus, a new paradigm is emerging in the field of oncoimmunology. Can GZMs act as protumoral factors under some circumstances? We review the diverse roles of GZMs in cancer progression, and new therapeutic opportunities emerging from targeting these protumoral roles.


Assuntos
Granzimas/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Animais , Biomarcadores , Progressão da Doença , Suscetibilidade a Doenças , Matriz Extracelular/imunologia , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Granzimas/antagonistas & inibidores , Granzimas/genética , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Imunidade , Vigilância Imunológica , Imunomodulação , Ativação Linfocitária/imunologia , Terapia de Alvo Molecular , Células Mieloides/imunologia , Células Mieloides/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
10.
Cell Death Differ ; 24(4): 747-758, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28338658

RESUMO

We have found that granzyme B (GB)-induced apoptosis also requires reactive oxygen species resulting from the alteration of mitochondrial complex I. How GB, which does not possess a mitochondrial targeting sequence, enter this organelle is unknown. We show that GB enters the mitochondria independently of the translocase of the outer mitochondrial membrane complex, but requires instead Sam50, the central subunit of the sorting and assembly machinery that integrates outer membrane ß-barrel proteins. Moreover, GB breaches the inner membrane through Tim22, the metabolite carrier translocase pore, in a mitochondrial heat-shock protein 70 (mtHsp70)-dependent manner. Granzyme A (GA) and caspase-3 use a similar route to the mitochondria. Finally, preventing GB from entering the mitochondria either by mutating lysine 243 and arginine 244 or depleting Sam50 renders cells more resistant to GB-mediated reactive oxygen species and cell death. Similarly, Sam50 depletion protects cells from GA-, GM- and caspase-3-mediated cell death. Therefore, cytotoxic molecules enter the mitochondria to induce efficiently cell death through a noncanonical Sam50-, Tim22- and mtHsp70-dependent import pathway.


Assuntos
Apoptose , Granzimas/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Linhagem Celular Tumoral , Doxorrubicina/toxicidade , Complexo I de Transporte de Elétrons/metabolismo , Granzimas/antagonistas & inibidores , Granzimas/genética , Células HeLa , Humanos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Membranas Mitocondriais/metabolismo , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Valinomicina/toxicidade
11.
BMB Rep ; 49(10): 560-565, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27439606

RESUMO

Granzyme A (GzmA) was first identified as a cytotoxic T lymphocyte protease protein with limited tissue expression. A number of cellular proteins are known to be cleaved by GzmA, and its function is to induce apoptosis. Histones H1, H2B, and H3 were identified as GzmA substrates during apoptotic cell death. Here, we demonstrated that histone H4 was cleaved by GzmA during staurosporine-induced cell death; however, in the presence of caspase inhibitors, staurosporine-treated Raji cells underwent necroptosis instead of apoptosis. Furthermore, histone H4 cleavage was blocked by the GzmA inhibitor nafamostat mesylate and by GzmA knockdown using siRNA. These results suggest that histone H4 is a novel substrate for GzmA in staurosporine-induced cells. [BMB Reports 2016; 49(10): 560-565].


Assuntos
Apoptose/efeitos dos fármacos , Granzimas/metabolismo , Histonas/metabolismo , Estaurosporina/toxicidade , Clorometilcetonas de Aminoácidos/farmacologia , Benzamidinas , Western Blotting , Inibidores de Caspase/farmacologia , Linhagem Celular Tumoral , Citometria de Fluxo , Granzimas/antagonistas & inibidores , Granzimas/genética , Guanidinas/farmacologia , Células HEK293 , Células HeLa , Humanos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Estaurosporina/farmacologia
12.
Int J Oncol ; 49(1): 352-60, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27121069

RESUMO

Breast cancer stem cells seem to play important roles in breast tumor recurrence and endocrine therapy resistance, although the underlying mechanisms have not been well established. Moreover, in some tumor systems the immunosurveillance failure against cancer cells has been related to the presence of the granzyme B inhibitor PI-9. This study explored the status of PI-9 in tumorspheres isolated from estrogen receptor-α positive (ERα+) breast cancer MCF7 cells. Studies were performed in tertiary tumorspheres which possess high levels of stemness markers (Nanog, Oct3/4 and Sox2) and self-renewal ability. The exposure to estrogens (17-ß estradiol and genistein) increased the number and sizes of tumorspheres, promoting cell proliferation as demonstrated by the increase in the proliferating cell nuclear antigen (PCNA). The study of the three isoforms (66, 46 and 36 kDa) of ERα disclosed that tertiary tumorspheres exhibit a marked increase in ERα36, while the level of ERα66, which is highly expressed in MCF7 cells, declines. Although it is known that PI-9 is a transcriptional target of ERα66, surprisingly in tertiary tumorspheres, despite the reduced level of ERα66, the protein and mRNA content of PI-9 is higher than in MCF7 cells. Treatment with estrogens further increased PI-9 level while decreased that of ERα66 isoform thus excluding the involvement of this receptor isoform in the event. Moreover, our studies also provided evidence that tertiary tumorspheres express elevated levels of CXCR4 and phospho-p38, suggesting that the high PI-9 content might be ascribed to the activation of the proliferative CXCR4/phospho-p38 axis. Taken together, these events could supply a selective advantage to breast cancer stem cells by interfering with immunosurveillance systems and open up the avenue to new possible targets for breast cancer treatment.


Assuntos
Neoplasias da Mama/genética , Receptor alfa de Estrogênio/biossíntese , Recidiva Local de Neoplasia/genética , Células-Tronco Neoplásicas/metabolismo , Serpinas/biossíntese , Neoplasias da Mama/patologia , Proliferação de Células/genética , Receptor alfa de Estrogênio/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genisteína/administração & dosagem , Granzimas/antagonistas & inibidores , Humanos , Células MCF-7 , Recidiva Local de Neoplasia/fisiopatologia , Células-Tronco Neoplásicas/patologia , Receptores CXCR4/biossíntese , Serpinas/genética , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
13.
FEBS J ; 283(9): 1734-47, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26936634

RESUMO

Granzymes are a family of serine proteases that were once thought to function exclusively as mediators of cytotoxic lymphocyte-induced target cell death. However, non-apoptotic roles for granzymes, including granzyme K (GzK), have been proposed. As recent studies have observed elevated levels of GzK in the plasma of patients diagnosed with clinical sepsis, we hypothesized that extracellular GzK induces a proinflammatory response in endothelial cells. In the present study, extracellular GzK proteolytically activated protease-activated receptor-1 leading to increased interleukin 6 and monocyte chemotactic protein 1 production in endothelial cells. Enhanced expression of intercellular adhesion molecule 1 along with an increased capacity for adherence of THP-1 cells was also observed. Characterization of downstream pathways implicated the mitogen-activated protein kinase p38 pathway for intercellular adhesion molecule 1 expression, and both the p38 and the extracellular signal-regulated protein kinases 1 and 2 pathways in cytokine production. GzK also increased tumour necrosis factor α-induced inflammatory adhesion molecule expression. Furthermore, the physiological inhibitor of GzK, inter-α-inhibitor protein, significantly inhibited GzK activity in vitro. In summary, extracellular GzK promotes a proinflammatory response in endothelial cells.


Assuntos
Granzimas/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Receptor PAR-1/genética , alfa-Globulinas/farmacologia , Butadienos/farmacologia , Adesão Celular , Linhagem Celular , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Regulação da Expressão Gênica , Granzimas/antagonistas & inibidores , Granzimas/genética , Granzimas/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Imidazóis/farmacologia , Interleucina-6/genética , Interleucina-6/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Monócitos/citologia , Monócitos/metabolismo , Nitrilas/farmacologia , Proteólise/efeitos dos fármacos , Piridinas/farmacologia , Receptor PAR-1/antagonistas & inibidores , Receptor PAR-1/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
PLoS One ; 11(3): e0151465, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26963506

RESUMO

BACKGROUND: Central nervous system (CNS) primitive neuroectodermal tumors (PNETs) are malignant primary brain tumors that occur in young infants. Using current standard therapy, up to 80% of the children still dies from recurrent disease. Cellular immunotherapy might be key to improve overall survival. To achieve efficient killing of tumor cells, however, immunotherapy has to overcome cancer-associated strategies to evade the cytotoxic immune response. Whether CNS-PNETs can evade the immune response remains unknown. METHODS: We examined by immunohistochemistry the immune response and immune evasion strategies in pediatric CNS-PNETs. RESULTS: Here, we show that CD4+, CD8+, γδ-T-cells, and Tregs can infiltrate pediatric CNS-PNETs, although the activation status of cytotoxic cells is variable. Pediatric CNS-PNETs evade immune recognition by downregulating cell surface MHC-I and CD1d expression. Intriguingly, expression of SERPINB9, SERPINB1, and SERPINB4 is acquired during tumorigenesis in 29%, 29%, and 57% of the tumors, respectively. CONCLUSION: We show for the first time that brain tumors express direct granzyme inhibitors (serpins) as a potential mechanism to overcome cellular cytotoxicity, which may have consequences for cellular immunotherapy.


Assuntos
Neoplasias Encefálicas/imunologia , Proteínas de Neoplasias/imunologia , Tumores Neuroectodérmicos Primitivos/imunologia , Inibidores de Serina Proteinase/imunologia , Serpinas/imunologia , Evasão Tumoral , Adolescente , Antígenos CD1d/imunologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Criança , Pré-Escolar , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Granzimas/antagonistas & inibidores , Granzimas/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Masculino , Tumores Neuroectodérmicos Primitivos/patologia , Tumores Neuroectodérmicos Primitivos/terapia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia
15.
Clin Exp Immunol ; 185(1): 107-18, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26990855

RESUMO

Besides mobilizing stem cells into the periphery, granulocyte colony-stimulating factor (G-CSF) has been shown to influence various types of innate and adaptive immune cells. For example, it impairs the effector function of cytotoxic T lymphocytes (CTLs). It is assumed that this effect is mediated indirectly by monocytes, regulatory T cells and immunomodulatory cytokines influenced by G-CSF. In this study, isolated G-CSF-treated CD8(+) T cells were stimulated antigen-dependently with peptide-major histocompatibility complex (pMHC)-coupled artificial antigen-presenting cells (aAPCs) or stimulated antigen-independently with anti-CD3/CD28 stimulator beads. By measuring the changes in interferon (IFN)-γ and granzyme B expression at the mRNA and protein level, we showed for the first time that G-CSF has a direct effect on CD8(+) CTLs, which was confirmed based on the reduced production of IFN-γ and granzyme B by the cytotoxic T cell line TALL-104 after G-CSF treatment. By investigating further elements affected by G-CSF in CTLs from stem cell donors and untreated controls, we found a decreased phosphorylation of extracellular-regulated kinase (ERK)1/2, lymphocyte-specific protein tyrosine kinase (Lck) and CD3ζ after G-CSF treatment. Additionally, miRNA-155 and activation marker expression levels were reduced. In summary, our results show that G-CSF directly influences the effector function of cytotoxic CD8(+) T cells and affects various elements of T cell activation.


Assuntos
Células Apresentadoras de Antígenos/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Antígenos CD28/genética , Antígenos CD28/imunologia , Complexo CD3/genética , Complexo CD3/imunologia , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/imunologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Regulação da Expressão Gênica , Granzimas/antagonistas & inibidores , Granzimas/genética , Granzimas/imunologia , Humanos , Interferon gama/antagonistas & inibidores , Interferon gama/genética , Interferon gama/imunologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/genética , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/imunologia , MicroRNAs/genética , MicroRNAs/imunologia , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/imunologia , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/imunologia , Cultura Primária de Células , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Transdução de Sinais , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia
16.
Oncotarget ; 7(8): 9368-87, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26824186

RESUMO

miR-143 and miR-145 are downregulated in colon cancer. Here, we tested the effect of restoring these miRNAs on sensitization to cetuximab in mutant KRAS (HCT116 and SW480) and wild-type KRAS (SW48) colon cancer cells. We evaluated cetuximab-mediated antibody-dependent cellular cytotoxicity (ADCC) and the modulation of signaling pathways involved in immune effector cell-mediated elimination of cancer cells. Stable miR-143 or miR-145 overexpression increased cell sensitivity to cetuximab, resulting in a significant increase of cetuximab-mediated ADCC independently of KRAS status. Importantly, HCT116 cells overexpressing these miRNAs triggered apoptosis in result of cetuximab-mediated ADCC, effected by peripheral blood mononuclear cells (p < 0.01). This was associated with increased apoptosis and caspase-3/7 activity, and reduced Bcl-2 protein expression (p < 0.01). In addition, caspase inhibition abrogated cetuximab-mediated ADCC in HCT116 cells overexpressing either miR-143 or miR-145 (p < 0.01). Furthermore, Bcl-2 silencing led to high level of cetuximab-mediated ADCC, compared to control siRNA (p < 0.05). Importantly, granzyme B inhibition, abrogated cetuximab-mediated ADCC, reducing caspase-3/7 activity (p < 0.01). Collectively, our data suggests that re-introduction of miR-143 or miR-145 may provide a new approach for development of therapeutic strategies to re-sensitize colon cancer cells to cetuximab by stimulating cetuximab-dependent ADCC to induce cell death.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Cetuximab/farmacologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , MicroRNAs/genética , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Genes bcl-2/genética , Granzimas/antagonistas & inibidores , Células HCT116 , Humanos , MicroRNAs/biossíntese , Proteínas Proto-Oncogênicas p21(ras)/genética , Interferência de RNA , RNA Interferente Pequeno/genética
17.
J Biol Chem ; 291(7): 3626-38, 2016 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-26670609

RESUMO

The intracellular protease inhibitor Sb9 (SerpinB9) is a regulator of the cytotoxic lymphocyte protease GzmB (granzyme B). Although GzmB is primarily involved in the destruction of compromised cells, recent evidence suggests that it is also involved in lysosome-mediated death of the cytotoxic lymphocyte itself. Sb9 protects the cell from GzmB released from lysosomes into the cytosol. Here we show that reactive oxygen species (ROS) generated within cytotoxic lymphocytes by receptor stimulation are required for lyososomal permeabilization and release of GzmB into the cytosol. Importantly, ROS also inactivate Sb9 by oxidizing a highly conserved cysteine pair (P1-P1' in rodents and P1'-P2' in other mammals) in the reactive center loop to form a vicinal disulfide bond. Replacement of the P4-P3' reactive center loop residues of the prototype serpin, SERPINA1, with the P4-P5' residues of Sb9 containing the cysteine pair is sufficient to convert SERPINA1 into a ROS-sensitive GzmB inhibitor. Conversion of the cysteine pair to serines in either human or mouse Sb9 results in a functional serpin that inhibits GzmB and resists ROS inactivation. We conclude that ROS sensitivity of Sb9 allows the threshold for GzmB-mediated suicide to be lowered, as part of a conserved post-translational homeostatic mechanism regulating lymphocyte numbers or activity. It follows, for example, that antioxidants may improve NK cell viability in adoptive immunotherapy applications by stabilizing Sb9.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Granzimas/metabolismo , Células Matadoras Naturais/metabolismo , Proteínas de Membrana/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Serpinas/metabolismo , Animais , Apoptose , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Células Cultivadas , Cistina/química , Granzimas/antagonistas & inibidores , Granzimas/química , Granzimas/genética , Humanos , Células Jurkat , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Lisossomos/enzimologia , Lisossomos/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mutantes , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Serpinas/química , Serpinas/genética
18.
J Immunol ; 194(6): 2786-95, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25667415

RESUMO

FcγRs are critical mediators of mAb cancer therapies, because they drive cytotoxic processes upon binding of effector cells to opsonized targets. Along with NK cells, monocytes are also known to destroy Ab-coated targets via Ab-dependent cellular cytotoxicity (ADCC). However, the precise mechanisms by which monocytes carry out this function have remained elusive. In this article, we show that human monocytes produce the protease granzyme B upon both FcγR and TLR8 activation. Treatment with TLR8 agonists elicited granzyme B and also enhanced FcγR-mediated granzyme B production in an additive fashion. Furthermore, monocyte-mediated ADCC against cetuximab-coated tumor targets was enhanced by TLR8 agonist treatment, and this enhancement of ADCC required granzyme B. Hence we have identified granzyme B as an important mediator of FcγR function in human monocytes and have uncovered another mechanism by which TLR8 agonists may enhance FcγR-based therapies.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Granzimas/metabolismo , Monócitos/metabolismo , Receptor 8 Toll-Like/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Angiotensinogênio/genética , Angiotensinogênio/metabolismo , Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Western Blotting , Células Cultivadas , Cetuximab , Análise por Conglomerados , Relação Dose-Resposta a Droga , Granzimas/antagonistas & inibidores , Granzimas/genética , Humanos , Imidazóis/farmacologia , Interleucina-2/genética , Interleucina-2/metabolismo , Monócitos/citologia , Monócitos/efeitos dos fármacos , NF-kappa B/genética , NF-kappa B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Perforina/genética , Perforina/metabolismo , Quinolinas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tiazóis/farmacologia , Fatores de Tempo , Receptor 8 Toll-Like/agonistas , Transcriptoma/efeitos dos fármacos
19.
Proteins ; 83(7): 1209-24, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25143259

RESUMO

Off-target binding connotes the binding of a small molecule of therapeutic significance to a protein target in addition to the primary target for which it was proposed. Progressively such off-targeting is emerging to be regular practice to reveal side effects. Chymase is an enzyme of hydrolase class that catalyzes hydrolysis of peptide bonds. A link between heart failure and chymase is ascribed, and a chymase inhibitor is in clinical phase II for treatment of heart failure. However, the underlying mechanisms of the off-target effects of human chymase inhibitors are still unclear. Here, we develop a robust computational strategy that is applicable to any enzyme system and that allows the prediction of drug effects on biological processes. Putative off-targets for chymase inhibitors were identified through various structural and functional similarity analyses along with molecular docking studies. Finally, literature survey was performed to incorporate these off-targets into biological pathways and to establish links between pathways and particular adverse effects. Off-targets of chymase inhibitors are linked to various biological pathways such as classical and lectin pathways of complement system, intrinsic and extrinsic pathways of coagulation cascade, and fibrinolytic system. Tissue kallikreins, granzyme M, neutrophil elastase, and mesotrypsin are also identified as off-targets. These off-targets and their associated pathways are elucidated for the effects of inflammation, cancer, hemorrhage, thrombosis, and central nervous system diseases (Alzheimer's disease). Prospectively, our approach is helpful not only to better understand the mechanisms of chymase inhibitors but also for drug repurposing exercises to find novel uses for these inhibitors.


Assuntos
Quimases/antagonistas & inibidores , Inibidores Enzimáticos/química , Simulação de Acoplamento Molecular , Bibliotecas de Moléculas Pequenas/química , Biologia de Sistemas/métodos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Doença de Alzheimer/patologia , Coagulação Sanguínea/efeitos dos fármacos , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/enzimologia , Doenças Cardiovasculares/patologia , Quimases/química , Quimases/metabolismo , Lectina de Ligação a Manose da Via do Complemento/efeitos dos fármacos , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Fibrinólise/efeitos dos fármacos , Granzimas/antagonistas & inibidores , Granzimas/química , Granzimas/metabolismo , Humanos , Elastase de Leucócito/antagonistas & inibidores , Elastase de Leucócito/química , Elastase de Leucócito/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Relação Estrutura-Atividade , Calicreínas Teciduais/antagonistas & inibidores , Calicreínas Teciduais/química , Calicreínas Teciduais/metabolismo , Tripsina/química , Tripsina/metabolismo , Interface Usuário-Computador
20.
J Immunol ; 194(1): 418-28, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25404359

RESUMO

Granzyme B (GzmB) plays a major role in CTLs and NK cell-mediated elimination of virus-infected cells and tumors. Human GzmB preferentially induces target cell apoptosis by cleaving the proapoptotic Bcl-2 family member Bid, which, together with Bax, induces mitochondrial outer membrane permeabilization. We previously showed that GzmB also induces a rapid accumulation of the tumor-suppressor protein p53 within target cells, which seems to be involved in GzmB-induced apoptosis. In this article, we show that GzmB-activated p53 accumulates on target cell mitochondria and interacts with Bcl-2. This interaction prevents Bcl-2 inhibitory effect on both Bax and GzmB-truncated Bid, and promotes GzmB-induced mitochondrial outer membrane permeabilization. Consequently, blocking p53-Bcl-2 interaction decreases GzmB-induced Bax activation, cytochrome c release from mitochondria, and subsequent effector caspases activation leading to a decreased sensitivity of target cells to both GzmB and CTL/NK-mediated cell death. Together, our results define p53 as a new important player in the GzmB apoptotic signaling pathway and in CTL/NK-induced apoptosis.


Assuntos
Apoptose/imunologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Granzimas/metabolismo , Linfócitos T Citotóxicos/imunologia , Proteína Supressora de Tumor p53/metabolismo , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/genética , Benzotiazóis/farmacologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Citocromos c/metabolismo , Ativação Enzimática , Granzimas/antagonistas & inibidores , Granzimas/farmacologia , Humanos , Células Matadoras Naturais/imunologia , Células MCF-7 , Mitocôndrias/imunologia , Membranas Mitocondriais/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Tolueno/análogos & derivados , Tolueno/farmacologia , Proteína Supressora de Tumor p53/genética , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA