Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
2.
Int Immunopharmacol ; 125(Pt B): 111226, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37976597

RESUMEN

OBJECTIVE: This study aimed to investigate the underlying regulatory effects of methionine enkephalin (MENK) on osteosarcoma. METHODS: The Cell Counting Kit-8 assay, clone formation, wound healing, transwell assay, and flow cytometry were performed to measure the effects of MENK on the proliferation, migration, invasion, and apoptosis of MG-63 and Saos-2 cells. Opiate growth factor receptor expression (OGFr) in cells was stably knocked down using siRNA. A tumor model was established by inoculating MG-63 cells into mice. Flow cytometry was performed to identify alterations in mice bone marrow, spleen, and tumor tissue immune cells. The phenotype of tumor-associated macrophages was determined using immunohistochemistry. After OGFr knockdown or/and treatment with MENK, Bax, Bcl-2, caspase 3, caspase 9, and PARP expression levels were characterized using qRT-PCR, western blot, and WES, respectively. RESULTS: MENK could significantly inhibit the proliferation, invasion, and migration of MG-63 and Saos-2, arrest the cell cycle in the G0/G1 phase, upregulate Bax, caspase 3, caspase 9, and PARP expression, and downregulate Bcl-2 expression. Tumor size and weight were lower in the MENK group than those in the control group. MENK-treated mice exhibited a reduced ratio of CD11b + Gr-1 + myeloid-derived suppressor cells. MENK increased the ratio of M1-type macrophages and decreased the proportion of M2-type macrophages in tumor tissue. Furthermore, the level of TNF-α significantly increased while that of IL-10 decreased in MENK-treated mice. The effect of MENK could be partly reversed by OGFr knockdown. CONCLUSION: MENK reduces the abundance of myeloid-derived suppressor cells, induces M1 polarization of macrophages, and exhibits an inhibitory effect on osteosarcoma.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Animales , Ratones , Caspasa 3 , Caspasa 9 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Proteína X Asociada a bcl-2 , Osteosarcoma/tratamiento farmacológico , Encefalina Metionina/farmacología , Encefalina Metionina/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico
3.
BMC Immunol ; 24(1): 38, 2023 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-37828468

RESUMEN

Novel prophylactic drugs and vaccination strategies for protection against influenza virus should induce specific effector T-cell immune responses in pulmonary airways and peripheral lymphoid organs. Designing approaches that promote T-cell-mediated responses and memory T-cell differentiation would strengthen host resistance to respiratory infectious diseases. The results of this study showed that pulmonary delivery of MENK via intranasal administration reduced viral titres, upregulated opioid receptor MOR and DOR, increased the proportions of T-cell subsets including CD8+ T cells, CD8+ TEM cells, NP/PA-effector CD8+ TEM cells in bronchoalveolar lavage fluid and lungs, and CD4+/CD8+ TCM cells in lymph nodes to protect mice against influenza viral challenge. Furthermore, we demonstrated that, on the 10th day of infection, the proportions of CD4+ TM and CD8+ TM cells were significantly increased, which meant that a stable TCM and TEM lineage was established in the early stage of influenza infection. Collectively, our data suggested that MENK administered intranasally, similar to the route of natural infection by influenza A virus, could exert antiviral activity through upregulating T-cell-mediated adaptive immune responses against influenza virus.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Ratones , Animales , Humanos , Linfocitos T CD8-positivos , Encefalina Metionina/farmacología , Células T de Memoria , Memoria Inmunológica , Ratones Endogámicos C57BL
4.
Int Immunopharmacol ; 124(Pt B): 110967, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37741126

RESUMEN

This study was to study the role of methionine enkephalin (menk) in cell invasion and migration as well as NK cells activation of tumor microenvironment in cervical cancer. The results showed that menk inhibited cervical cancer migration and invasion. In addition, we found menk affected epithelial to mesenchymal transition (EMT) related indicators, with increasing E-cadherin level, decreasing N-cadherin and vimentin level. Through in vivo mouse model, we found that menk IFNγ and NKP46 expression was upregulated in tumor tissues by menk compared with controls, while LAG3 expression was inhibited by menk, besides, there was an upregulation of CD11b+ NCR1+ NKs of tumor microenvironment in cervical cancer. Therefore, we concluded that menk inhibited cancer migration and invasion via affecting EMT related indicators and activated CD11b+ NCR1+ NKs of tumor microenvironment in cervical cancer, laying a theoretical foundation for the further clinical treatment of menk.


Asunto(s)
Neoplasias del Cuello Uterino , Humanos , Femenino , Ratones , Animales , Neoplasias del Cuello Uterino/tratamiento farmacológico , Encefalina Metionina/farmacología , Transición Epitelial-Mesenquimal , Microambiente Tumoral , Receptor 1 Gatillante de la Citotoxidad Natural , Línea Celular Tumoral , Movimiento Celular
5.
Int Immunopharmacol ; 118: 110064, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36989897

RESUMEN

Metastasis is one of the most difficult challenges for clinical lung cancer treatment. Epithelial-mesenchymal transition (EMT) is the crucial step of tumor metastasis. Immune cells in the tumor microenvironment (TME), such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), promote cancer cell EMT. In this study, we explored the effect of methionine enkephalin (MENK) on the EMT process in vitro and in vivo, and its influence on TAMs, MDSCs, and associated cytokines in vivo. The results showed that MENK suppressed growth, migration, and invasion of lung cancer cells and inhibited the EMT process by interacting with opioid growth factor receptor. MENK reduced the number of M2 macrophages and MDSC infiltration, and downregulated the expression of interleukin-10 and transforming growth factor-ß1 in both primary and metastatic tumors of nude mice. The present findings suggest that MENK is a potential target for suppressing metastasis in lung cancer treatment.


Asunto(s)
Neoplasias Pulmonares , Células Supresoras de Origen Mieloide , Animales , Ratones , Transición Epitelial-Mesenquimal , Macrófagos Asociados a Tumores/metabolismo , Encefalina Metionina/farmacología , Encefalina Metionina/uso terapéutico , Encefalina Metionina/metabolismo , Microambiente Tumoral , Ratones Desnudos , Línea Celular Tumoral , Movimiento Celular
6.
Int Immunopharmacol ; 111: 109125, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35988519

RESUMEN

There is evidence that methionine enkephalin (MENK), an opioid peptide, promotes anti-tumor immune responses. In this study, the effect of MENK on colorectal cancer (CRC) and its mechanisms of action were examined in vivo. The intraperitoneal administration of 20 mg/kg MENK effectively inhibited MC38 subcutaneous colorectal tumor growth in mice. MENK inhibited tumor progression by increasing the immunogenicity and recognition of MC38 cells. MENK down-regulated the oncogene Kras and anti-apoptotic Bclxl and Bcl2, suppressed Il1b, Il6, iNOS, and Arg1 (encoding inflammatory cytokines), and increased Il17a and Il10 levels. MENK promoted a tumor suppressive state by decreasing the immune checkpoints Pd-1, Pd-l1, Lag3, Flgl1, and 2b4 in CRC. MENK also altered the immune status of the tumor immune microenvironment (TIME). It increased the infiltration of M1-type macrophages, CD8+T cells, and CD4+T cells and decreased the proportions of G-MDSCs, M-MDSCs, and M2-type macrophages. MENK accelerated CD4+TEM and CD8+TEM cell activation in the TIME and up-regulated IFN-γ, TNF-α, and IL-17A in CD4+T cells and Granzyme B in CD8+T cells. In addition, analyses of PD-1 and PD-L1 expression indicated that MENK promoted the anti-tumor immune response mediated by effector T cells. Finally, OGFr was up-regulated at the protein and mRNA levels by MENK, and the inhibitory effects of MENK on tumor growth were blocked by NTX, a specific blocker of OGFr. These finding indicate that MENK remodels the TIME in CRC to inhibit tumor progression by binding to OGFr. MENK is a potential therapeutic agent for CRC, especially for improving the efficacy of immunotherapy.


Asunto(s)
Neoplasias Colorrectales , Encefalina Metionina , Animales , Antígeno B7-H1 , Neoplasias Colorrectales/tratamiento farmacológico , Encefalina Metionina/farmacología , Encefalina Metionina/uso terapéutico , Factores Inmunológicos , Ratones , Receptor de Muerte Celular Programada 1 , Microambiente Tumoral
7.
Int Immunopharmacol ; 110: 108933, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35738090

RESUMEN

Immunotherapy for cervical carcinoma is becoming increasingly important recently. In these studies methionine enkephalin (menk) is shown to inhibit cervical tumor cell proliferation in vitro in association with an increase in the expression of apoptosis markers and mediators, including an increase in fas, caspase 8, and caspase 3 expression and intrinsic expression of the signaling pathway mediator bax. In vivo, tumor growth was restrained in mice xenotransplant model with typical pathological features of apoptosis. Furthermore, myeloid derived suppressor cells (MDSCs) had a significant decrease in circulation and in tumor site. In brief, these findings showed menk could inhibit tumor growth in vitro and in vivo, providing direction of further research and clinical application prospect.


Asunto(s)
Carcinoma , Células Supresoras de Origen Mieloide , Neoplasias del Cuello Uterino , Animales , Apoptosis , Línea Celular Tumoral , Encefalina Metionina/metabolismo , Encefalina Metionina/farmacología , Encefalina Metionina/uso terapéutico , Femenino , Humanos , Factores Inmunológicos/metabolismo , Ratones , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/metabolismo
9.
Int Immunopharmacol ; 108: 108881, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35623292

RESUMEN

Interleukin (IL)-10 family cytokines include IL-10, IL-19, IL-20, IL-22, IL-24, IL-26, IL-28A, IL-28B, and IL-29. These cytokines play crucial regulatory roles in various biological reactions and diseases. In recent years, several studies have shown that the IL-10 family plays a vital role in bone and joint diseases, including bone metabolic diseases, fractures, osteoarthritis, rheumatoid arthritis, and bone tumors. Herein, the recent progress on the regulatory role of IL-10 family of cytokines in the occurrence and development of bone and joint diseases has been summarized. This review will provide novel directions for immunotherapy of bone and joint diseases.


Asunto(s)
Artritis Reumatoide , Osteoartritis , Artritis Reumatoide/metabolismo , Biología , Citocinas , Humanos , Interleucina-10
10.
Int Immunopharmacol ; 103: 108484, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34954558

RESUMEN

Cervical cancer is one of the most common in the female genital tract and remains a leading cause that threatens the health and lives of women worldwide, although preventive vaccines and early diagnosis have reduced mortality. While treatment by operation and chemoradiotherapy for early-stage patients achieve good outcomes, the great majority of cervical cancers caused by the human papilloma virus (HPV) make immunotherapy realizable for patients with advanced and recurrent cervical cancer. To date, some clinical trials of checkpoint immunotherapy in cervical cancer have indicated significant benefits of programmed cell death-1/programmed cell death-ligand 1 (PD-1/PD-L1) inhibitors, providing strong evidence for PD-1/PD-L1 as a therapeutic target. In this review article, we discuss the role of PD-L1 and the application of PD-L1 inhibitors in cervical cancer, with the aim of providing direction for future research.


Asunto(s)
Antígeno B7-H1 , Biomarcadores de Tumor , Neoplasias del Cuello Uterino , Antígeno B7-H1/metabolismo , Biomarcadores de Tumor/metabolismo , Femenino , Humanos , Inmunoterapia , Neoplasias Pulmonares , Recurrencia Local de Neoplasia , Pronóstico , Receptor de Muerte Celular Programada 1 , Neoplasias del Cuello Uterino/diagnóstico , Neoplasias del Cuello Uterino/tratamiento farmacológico
11.
Biomed Res Int ; 2021: 8851986, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34805408

RESUMEN

OBJECTIVE: Natural killer (NK) cell-deficient mice are useful models in biomedical research. NOD/SCID mice have been used as a model of this type in research. However, the actual status of NK cells in NOD/SCID mice and CB17/SCID mice in comparison with that in BALB/c mice has not been sufficiently evaluated. METHODS: Splenocytes from naïve or poly(I:C)-treated mice were isolated for phenotyping and analysis of cytotoxicity-related molecules and inhibitory receptors; for cytotoxicity assay, purified NK cells were also used. RESULTS: The proportion of splenic NK cells did not differ significantly between NOD/SCID and CB17/SCID mice. The perforin levels in NK cells were similar between the poly(I:C)-treated CB17/SCID and NOD/SCID mice, while the granzyme B and NKG2A/C/E levels in NK cells from NOD/SCID mice were significantly lower than those from CB17/SCID mice. Moreover, the NKG2D and Ly49A levels in NK cells from NOD/SCID mice were higher than those from CB17/SCID. The splenocytes from CB17/SCID mice showed higher cytotoxicity than those from NOD/SCID mice, while the cytotoxicity of purified NK cells basically did not differ between the two strains. After in vitro stimulation with cytokines, the splenocytes from CB17/SCID mice showed higher IFN-γ production than those from NOD/SCID mice; however, NK cells did not. CONCLUSION: There was no significant difference in the proportion of splenic NK cells between CB17/SCID and NOD/SCID mice, and the function of NK cells was only partially compromised in NOD/SCID mice. Caution should be taken when considering the use of NOD/SCID mice as an NK-deficient model.


Asunto(s)
Células Asesinas Naturales/inmunología , Ratones Endogámicos NOD/inmunología , Ratones SCID/inmunología , Animales , Antígenos Ly/metabolismo , Citocinas/biosíntesis , Citotoxicidad Inmunológica , Inmunidad Innata , Técnicas In Vitro , Células Asesinas Naturales/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Animales , Modelos Inmunológicos , Subfamilia B de Receptores Similares a Lectina de Células NK/metabolismo , Receptores Similares a Lectina de Células NK/metabolismo , Especificidad de la Especie
13.
Int Immunopharmacol ; 99: 107713, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34426103

RESUMEN

Methionine enkephalin (MENK) has an important role in both neuroendocrine and immune systems. MENK was known as an opioid growth factor (OGF) for its growth regulatory characteristics. OGF interacts with the OGF receptor (OGFr) to inhibit DNA synthesis by upregulating p16 and/or p21, which delays the cell cycle transition from G0/G1 to S phase, and inhibits cell proliferation. In addition, OGF combines with OGFr in immune cells to exert its immunomodulatory activity and regulate immune function. OGF has been studied as an immunomodulator in a variety of autoimmune diseases, including multiple sclerosis, inflammatory bowel disease, diabetes and viral infections, and has been proven to relieve symptoms of certain diseases in animal and in vitro experiments. Also, OGF and OGFr have various anti-tumor molecular mechanisms. OGF can be used as the primary therapy alone or combined with other drugs to treat tumors. This article summarizes the research progress of OGF in immune-related diseases and cancer diseases.


Asunto(s)
Encefalina Metionina/metabolismo , Enfermedades del Sistema Inmune/metabolismo , Neoplasias/metabolismo , Animales , Humanos , Enfermedades del Sistema Inmune/tratamiento farmacológico , Antagonistas de Narcóticos/uso terapéutico , Neoplasias/tratamiento farmacológico
14.
Int Immunopharmacol ; 99: 108017, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34352568

RESUMEN

Colorectal cancer (CRC), known as a frequently fatal disease, ranking as the third most common malignancy, is the second leading cause of cancer related mortality worldwide. Metastases are common in CRC patients which account for approximately 25% of the patients at diagnosis, 50% of patients during treatment which is associated closely with CRC mortality. Conventional therapies such as surgery, chemotherapy, and radiotherapy are standards of care for the treatment of CRC patients. However, primary tumor recurrence and secondary disease in patients receiving standard of care treatment modalities occur in 50% of patients so that new treatment modalities are needed. Immune checkpoint inhibition (ICI) has transformed the management of patients suffered from metastatic CRC (mCRC) with mismatch repair deficiency (dMMR) and microsatellite instability (MSI) -high (MSI-H) while manifests ineffectiveness in preserved mismatch repair (pMMR) or microsatellite stable (MSS) "cold" tumors which makes up the majority (95%) of mCRC. In this review, we mainly lay emphasis on the development of combinations in therapy strategies with ICIs with other immune based treatment approaches to increase the intra-tumoral immune response and render tumors 'immune-reactive', thereby increasing the efficacy of tumor immunotherapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Quimioradioterapia/métodos , Neoplasias Colorrectales/terapia , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Quimioradioterapia/tendencias , Ensayos Clínicos como Asunto , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/mortalidad , Reparación de la Incompatibilidad de ADN , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/efectos de la radiación , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inestabilidad de Microsatélites , Supervivencia sin Progresión , Inhibidores de Proteínas Quinasas/farmacología
15.
Int Immunopharmacol ; 99: 107999, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34315116

RESUMEN

This study examined the antitumor effect of methionine enkephalin (MENK) against lung cancer in vivo and in vitro and explored the underlying mechanisms. Changes in the immune status of the tumor microenvironment (TME) in response to MENK administration were examined in mice. MENK significantly inhibited the proliferation of lung cancer cells in vivo and in vitro by regulating the Wnt/ß-catenin pathway and causing cell cycle arrest at the G0/G1 phase. Knockdown of opioid growth factor receptor abolished the effect of MENK on lung cancer cells. The immune status of the TME of mice differed between the MENK and control groups. MENK increased the infiltration of M1-type macrophages, natural killer cells, CD8+ T cells, CD4+ T cells, and dendritic cells into the TME, and decreased the proportion of myeloid inhibitory cells and M2-type macrophages. Immunohistochemical analysis of the expression of cytokines in the TME showed that MENK upregulated IL-15, IL-21, IFN-γ, and granzyme B and downregulated IL-10 and TGF-ß1 in mice. Taken together, these finding indicate that MENK may be a potential agent for lung cancer treatment in the future, especially for overcoming immune escape and immune resistance.


Asunto(s)
Encefalina Metionina/metabolismo , Encefalina Metionina/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocinas/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Encefalina Metionina/genética , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño , Linfocitos T/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos
16.
Int Immunopharmacol ; 99: 107996, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34311187

RESUMEN

The antitumor effects of methionine enkephalin (MENK), also known as opioid growth factor (OGF), including its inhibitory effects on cutaneous squamous cell carcinoma (CSCC), have been established. In this study, we determined the precise mechanism by which MENK suppresses CSCC cell growth. In particular, MENK induced G0/G1 cell cycle arrest and promoted apoptosis in CSCC cells via the Bcl-2/Bax/Caspase-3 signaling pathway. Moreover, MENK reduced immunosuppression by downregulating the number of myeloid-derived suppressor cells (MDSCs) and regulating the polarization of tumor-associated macrophages from M2 to M1 in vivo. Furthermore, JAK2/STAT3, an important tumor-promotion and immunosuppression signaling pathway that is involved in MDSC expansion in tumors and macrophage polarization, was inhibited. These findings highlight the potential of the JAK2/STAT3 signaling pathway as a therapeutic target and suggest the clinical application of MENK for CSCC.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Encefalina Metionina/metabolismo , Encefalina Metionina/farmacología , Macrófagos/efectos de los fármacos , Células Supresoras de Origen Mieloide/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Polaridad Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Encefalina Metionina/genética , Femenino , Humanos , Terapia de Inmunosupresión , Janus Quinasa 2/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Interferente Pequeño , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos
17.
Int Immunopharmacol ; 98: 107837, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34116288

RESUMEN

The aim of this study was to investigate how methionine enkephalin (MENK) regulates the biological behavior of lung cancer cells and to further explore its anti-lung cancer mechanisms in vitro and in vivo. The results showed that MENK enhanced the expression of opioid receptor (OGFr) and induced apoptosis of lung cancer cells by activating the Bcl-1/Bax/caspase-3 signaling pathway in vitro and in vivo. However, the regulatory effects of MENK disappeared after blockade of the OGFr. This confirmed that a prerequisite for the anti-tumor action of MENK is binding to OGFr. Additionally, we observed that MENK treatment enhanced the immunogenicity of lung cancer by enhancing the exposure of calreticulin and high mobility group box 1, and increasing the expression of NKG2D ligands. Further studies showed that MENK treatment increased the expression of natural killer (NK) cell-related cytokines such as granzyme B and interferon-γ and NK cell activation. Thus, we concluded that MENK might inhibit the proliferation of lung cancer cells by activating the Bcl-2/Bax/caspase-3 signaling pathway and enhancing immunogenicity and NK cell-driven tumor immunity.


Asunto(s)
Antineoplásicos/metabolismo , Caspasa 3/metabolismo , Encefalina Metionina/metabolismo , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Células A549 , Animales , Humanos , Inmunidad Celular , Neoplasias Pulmonares/inmunología , Ratones , Ratones Endogámicos C57BL , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Transducción de Señal
18.
Int Immunopharmacol ; 96: 107714, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33989971

RESUMEN

Naltrexone (NTX) is a nonspecific opioid antagonist that exerts pharmacological effects on the opioid axis by blocking opioid receptors distributed in cytoplastic and nuclear regions. NTX has been used in opioid use disorder (OUD), immune-associated diseases, alcoholism, obesity, and chronic pain for decades. However, low-dose naltrexone (LDN) also exhibits remarkable inhibition of DNA synthesis, viability, and other functions in numerous cancers and is involved in immune remodeling against tumor invasion and chemical toxicity. The potential anticancer activity of LDN is a focus of basic research. Herein, we summarize the associated studies on LDN oncotherapy to highlight the potential mechanisms and prospective clinical applications.


Asunto(s)
Naltrexona/farmacología , Neoplasias/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Humanos , Antagonistas de Narcóticos/farmacología , Neoplasias/metabolismo , Neoplasias/patología , Receptores Opioides/metabolismo
19.
Int Immunopharmacol ; 96: 107733, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33965882

RESUMEN

Cutaneous squamous cell carcinoma (CSCC) is a common skin tumour. Due to weak immunogenicity, recurrence is frequent after treatment. In this study, we explored the effects and mechanisms of methionine enkephalin (MENK), an endogenous opioid peptide and negative growth regulator, in CSCC. MENK inhibited A431 cell proliferation and promoted apoptosis through the opioid growth factor receptor (OGFr). Importantly, MENK also induced autophagy in CSCC and stimulated the emission of DAMPs in A431 cells, which resulted in enhanced activation of dendritic cells (DC).In conclusion, MENK provides an effective method with therapeutic potential to modulate the CSCC microenvironment by utilizing autophagy in the cancer cells.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Células Dendríticas/inmunología , Encefalina Metionina/farmacología , Antígenos de Histocompatibilidad Clase II/inmunología , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Autofagia/efectos de los fármacos , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular , Femenino , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Neurotransmisores/farmacología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Hematol Oncol ; 14(1): 71, 2021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33910591

RESUMEN

BACKGROUND: CD8+ T cell-mediated adaptive cellular immunity and natural killer (NK) cell-mediated innate immunity both play important roles in tumour immunity. This study aimed to develop therapeutic tumour vaccines based on double-activation of CD8+ T and NK cells. METHODS: The immune Epitope database, Molecular Operating Environment software, and enzyme-linked immunosorbent assay were used for epitope identification. Flow cytometry, confocal microscopy, UPLC-QTOF-MS, and RNA-seq were utilized for evaluating immunity of PBMC-derived DCs, CD8+ T or NK cells and related pathways. HLA-A2.1 transgenic mice combined with immunologically reconstituted tumour-bearing mice were used to examine the antitumour effect and safety of epitope vaccines. RESULTS: We identified novel HLA-A2.1-restricted extracellular matrix protein 1(ECM1)-derived immunodominant epitopes in which LA induced a potent immune response. We also found that LA-loaded DCs upregulated the frequency of CD3+/CD8+ T cells, CD45RO+/CD69+ activated memory T cells, and CD3-/CD16+/CD56+ NK cells. We demonstrated cytotoxic granule release of LA/DC-CTLs or LA/DC-NK cells and cytotoxicity against tumour cells and microtissue blocks via the predominant IFN-γ/perforin/granzyme B cell death pathway. Further investigating the mechanism of LA-mediated CD8+ T activation, we found that LA could be internalized into DCs through phagocytosis and then formed a LA-MHC-I complex presented onto the DC surface for recognition of the T cell receptor to upregulate Zap70 phosphorylation levels to further activate CD8+ T cells by DC-CTL interactions. In addition, LA-mediated DC-NK crosstalk through stimulation of the TLR4-p38 MAPK pathway increased MICA/B expression on DCs to interact with NKG2D for NK activation. Promisingly, LA could activate CD8+ T cells and NK cells simultaneously via interacting with DCs to suppress tumours in vivo. Moreover, the safety of LA was confirmed. CONCLUSIONS: LA-induced immune antitumour activity through DC cross-activation with CD8+ T and NK cells, which demonstrated proof-of-concept evidence for the capability and safety of a novel therapeutic tumour vaccine.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Proteínas de la Matriz Extracelular/inmunología , Antígeno HLA-A2/inmunología , Células Asesinas Naturales/inmunología , Neoplasias/terapia , Animales , Comunicación Celular/inmunología , Línea Celular Tumoral , Humanos , Epítopos Inmunodominantes/inmunología , Células MCF-7 , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Neoplasias/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA