Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.798
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 20(8): 1012-1022, 2019 08.
Article in English | MEDLINE | ID: mdl-31263276

ABSTRACT

The plasma membrane tetraspan molecule MS4A4A is selectively expressed by macrophage-lineage cells, but its function is unknown. Here we report that MS4A4A was restricted to murine and human mononuclear phagocytes and was induced during monocyte-to-macrophage differentiation in the presence of interleukin 4 or dexamethasone. Human MS4A4A was co-expressed with M2/M2-like molecules in subsets of normal tissue-resident macrophages, infiltrating macrophages from inflamed synovium and tumor-associated macrophages. MS4A4A interacted and colocalized with the ß-glucan receptor dectin-1 in lipid rafts. In response to dectin-1 ligands, Ms4a4a-deficient macrophages showed defective signaling and defective production of effector molecules. In experimental models of tumor progression and metastasis, Ms4a4a deficiency in macrophages had no impact on primary tumor growth, but was essential for dectin-1-mediated activation of macrophages and natural killer (NK) cell-mediated metastasis control. Thus, MS4A4A is a tetraspan molecule selectively expressed in macrophages during differentiation and polarization, essential for dectin-1-dependent activation of NK cell-mediated resistance to metastasis.


Subject(s)
Killer Cells, Natural/immunology , Lectins, C-Type/metabolism , Macrophages/immunology , Membrane Proteins/metabolism , Neoplasm Metastasis/immunology , Neoplasms/immunology , Animals , Cell Differentiation/immunology , Cell Lineage , Dexamethasone/pharmacology , Humans , Interleukin-4/metabolism , Lymphocyte Activation/immunology , Macrophage Activation/immunology , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Neoplasm Metastasis/prevention & control , Neoplasms/pathology
2.
Nature ; 632(8024): 411-418, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39048831

ABSTRACT

It is estimated that only 0.02% of disseminated tumour cells are able to seed overt metastases1. While this suggests the presence of environmental constraints to metastatic seeding, the landscape of host factors controlling this process remains largely unclear. Here, combining transposon technology2 and fluorescence niche labelling3, we developed an in vivo CRISPR activation screen to systematically investigate the interactions between hepatocytes and metastatic cells. We identify plexin B2 as a critical host-derived regulator of liver colonization in colorectal and pancreatic cancer and melanoma syngeneic mouse models. We dissect a mechanism through which plexin B2 interacts with class IV semaphorins on tumour cells, leading to KLF4 upregulation and thereby promoting the acquisition of epithelial traits. Our results highlight the essential role of signals from the liver parenchyma for the seeding of disseminated tumour cells before the establishment of a growth-promoting niche. Our findings further suggest that epithelialization is required for the adaptation of CRC metastases to their new tissue environment. Blocking the plexin-B2-semaphorin axis abolishes metastatic colonization of the liver and therefore represents a therapeutic strategy for the prevention of hepatic metastases. Finally, our screening approach, which evaluates host-derived extrinsic signals rather than tumour-intrinsic factors for their ability to promote metastatic seeding, is broadly applicable and lays a framework for the screening of environmental constraints to metastasis in other organs and cancer types.


Subject(s)
CRISPR-Cas Systems , Hepatocytes , Liver Neoplasms , Liver , Neoplasm Metastasis , Nerve Tissue Proteins , Animals , Female , Humans , Male , Mice , Cell Line, Tumor , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , CRISPR-Cas Systems/genetics , Disease Models, Animal , DNA Transposable Elements , Fluorescence , Hepatocytes/metabolism , Hepatocytes/cytology , Hepatocytes/pathology , Kruppel-Like Factor 4/metabolism , Liver/cytology , Liver/metabolism , Liver/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Melanoma/metabolism , Melanoma/pathology , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Semaphorins/antagonists & inhibitors , Semaphorins/metabolism
3.
Nature ; 611(7936): 603-613, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36352230

ABSTRACT

Around 30-40% of patients with colorectal cancer (CRC) undergoing curative resection of the primary tumour will develop metastases in the subsequent years1. Therapies to prevent disease relapse remain an unmet medical need. Here we uncover the identity and features of the residual tumour cells responsible for CRC relapse. An analysis of single-cell transcriptomes of samples from patients with CRC revealed that the majority of genes associated with a poor prognosis are expressed by a unique tumour cell population that we named high-relapse cells (HRCs). We established a human-like mouse model of microsatellite-stable CRC that undergoes metastatic relapse after surgical resection of the primary tumour. Residual HRCs occult in mouse livers after primary CRC surgery gave rise to multiple cell types over time, including LGR5+ stem-like tumour cells2-4, and caused overt metastatic disease. Using Emp1 (encoding epithelial membrane protein 1) as a marker gene for HRCs, we tracked and selectively eliminated this cell population. Genetic ablation of EMP1high cells prevented metastatic recurrence and mice remained disease-free after surgery. We also found that HRC-rich micrometastases were infiltrated with T cells, yet became progressively immune-excluded during outgrowth. Treatment with neoadjuvant immunotherapy eliminated residual metastatic cells and prevented mice from relapsing after surgery. Together, our findings reveal the cell-state dynamics of residual disease in CRC and anticipate that therapies targeting HRCs may help to avoid metastatic relapse.


Subject(s)
Colorectal Neoplasms , Neoplasm Metastasis , Neoplasm Proteins , Neoplasm Recurrence, Local , Neoplasm, Residual , Receptors, Cell Surface , Animals , Humans , Mice , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Disease Progression , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/prevention & control , Neoplasm Recurrence, Local/therapy , Neoplasm, Residual/genetics , Neoplasm, Residual/pathology , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Neoplasm Metastasis/therapy , Disease Models, Animal , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Neoadjuvant Therapy , Immunotherapy
4.
Nature ; 571(7763): 127-131, 2019 07.
Article in English | MEDLINE | ID: mdl-31243371

ABSTRACT

Cancer metastasis is the primary cause of morbidity and mortality, and accounts for up to 95% of cancer-related deaths1. Cancer cells often reprogram their metabolism to efficiently support cell proliferation and survival2,3. However, whether and how those metabolic alterations contribute to the migration of tumour cells remain largely unknown. UDP-glucose 6-dehydrogenase (UGDH) is a key enzyme in the uronic acid pathway, and converts UDP-glucose to UDP-glucuronic acid4. Here we show that, after activation of EGFR, UGDH is phosphorylated at tyrosine 473 in human lung cancer cells. Phosphorylated UGDH interacts with Hu antigen R (HuR) and converts UDP-glucose to UDP-glucuronic acid, which attenuates the UDP-glucose-mediated inhibition of the association of HuR with SNAI1 mRNA and therefore enhances the stability of SNAI1 mRNA. Increased production of SNAIL initiates the epithelial-mesenchymal transition, thus promoting the migration of tumour cells and lung cancer metastasis. In addition, phosphorylation of UGDH at tyrosine 473 correlates with metastatic recurrence and poor prognosis of patients with lung cancer. Our findings reveal a tumour-suppressive role of UDP-glucose in lung cancer metastasis and uncover a mechanism by which UGDH promotes tumour metastasis by increasing the stability of SNAI1 mRNA.


Subject(s)
Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/prevention & control , RNA Stability , Snail Family Transcription Factors/genetics , Uridine Diphosphate Glucose/metabolism , Animals , Cell Line, Tumor , Cell Movement , ELAV-Like Protein 1/deficiency , ELAV-Like Protein 1/genetics , ELAV-Like Protein 1/metabolism , Epithelial-Mesenchymal Transition , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Mice , Mice, Nude , Phosphotyrosine/metabolism , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Snail Family Transcription Factors/biosynthesis , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/genetics , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Uridine Diphosphate Glucuronic Acid/metabolism
5.
Proc Natl Acad Sci U S A ; 119(34): e2207841119, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35969778

ABSTRACT

The targeted delivery of messenger RNA (mRNA) to desired organs remains a great challenge for in vivo applications of mRNA technology. For mRNA vaccines, the targeted delivery to the lymph node (LN) is predicted to reduce side effects and increase the immune response. In this study, we explored an endogenously LN-targeting lipid nanoparticle (LNP) without the modification of any active targeting ligands for developing an mRNA cancer vaccine. The LNP named 113-O12B showed increased and specific expression in the LN compared with LNP formulated with ALC-0315, a synthetic lipid used in the COVID-19 vaccine Comirnaty. The targeted delivery of mRNA to the LN increased the CD8+ T cell response to the encoded full-length ovalbumin (OVA) model antigen. As a result, the protective and therapeutic effect of the OVA-encoding mRNA vaccine on the OVA-antigen-bearing B16F10 melanoma model was also improved. Moreover, 113-O12B encapsulated with TRP-2 peptide (TRP2180-188)-encoding mRNA also exhibited excellent tumor inhibition, with the complete response of 40% in the regular B16F10 tumor model when combined with anti-programmed death-1 (PD-1) therapy, revealing broad application of 113-O12B from protein to peptide antigens. All the treated mice showed long-term immune memory, hindering the occurrence of tumor metastatic nodules in the lung in the rechallenging experiments that followed. The enhanced antitumor efficacy of the LN-targeting LNP system shows great potential as a universal platform for the next generation of mRNA vaccines.


Subject(s)
Cancer Vaccines , Nanoparticles , Neoplasms , mRNA Vaccines , Amino Alcohols , Animals , Antigens/metabolism , CD8-Positive T-Lymphocytes , Cancer Vaccines/therapeutic use , Decanoates , Immunologic Memory , Liposomes , Lymph Nodes , Mice , Neoplasm Metastasis/prevention & control , Neoplasms/therapy , Ovalbumin , mRNA Vaccines/therapeutic use
6.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Article in English | MEDLINE | ID: mdl-35131941

ABSTRACT

Lysine-specific demethylase 6A (KDM6A), also named UTX, is frequently mutated in bladder cancer (BCa). Although known as a tumor suppressor, KDM6A's therapeutic potential in the metastasis of BCa remains elusive. It also remains difficult to fulfill the effective up-regulation of KDM6A levels in bladder tumor tissues in situ to verify its potential in treating BCa metastasis. Here, we report a mucoadhesive messenger RNA (mRNA) nanoparticle (NP) strategy for the intravesical delivery of KDM6A-mRNA in mice bearing orthotopic Kdm6a-null BCa and show evidence of KDM6A's therapeutic potential in inhibiting the metastasis of BCa. Through this mucoadhesive mRNA NP strategy, the exposure of KDM6A-mRNA to the in situ BCa tumors can be greatly prolonged for effective expression, and the penetration can be also enhanced by adhering to the bladder for sustained delivery. This mRNA NP strategy is also demonstrated to be effective for combination cancer therapy with other clinically approved drugs (e.g., elemene), which could further enhance therapeutic outcomes. Our findings not only report intravesical delivery of mRNA via a mucoadhesive mRNA NP strategy but also provide the proof-of-concept for the usefulness of these mRNA NPs as tools in both mechanistic understanding and translational study of bladder-related diseases.


Subject(s)
Histone Demethylases/pharmacology , Nanoparticles/chemistry , Neoplasm Metastasis/prevention & control , RNA, Messenger/pharmacology , Adhesiveness , Administration, Intravesical , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genetic Therapy , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Mice , Mice, Nude , Mucous Membrane , Neoplasms, Experimental/therapy , RNA, Messenger/administration & dosage , RNA, Messenger/metabolism , Urinary Bladder Neoplasms
7.
Traffic ; 23(7): 374-390, 2022 07.
Article in English | MEDLINE | ID: mdl-35575181

ABSTRACT

E-cadherin has a fundamental role in epithelial tissues by providing cell-cell adhesion. Polarised E-cadherin exocytosis to the lateral plasma membrane is central for cell polarity and epithelial homeostasis. Loss of E-cadherin secretion compromises tissue integrity and is a prerequisite for metastasis. Despite this pivotal role of E-cadherin secretion, the transport mechanism is still unknown. Here we identify Myosin V as the motor for E-cadherin secretion. Our data reveal that Myosin V and F-actin are required for the formation of a continuous apicolateral E-cadherin belt, the zonula adherens. We show by live imaging how Myosin V transports E-cadherin vesicles to the plasma membrane, and distinguish two distinct transport tracks: an apical actin network leading to the zonula adherens and parallel actin bundles leading to the basal-most region of the lateral membrane. E-cadherin secretion starts in endosomes, where Rab11 and Sec15 recruit Myosin V for transport to the zonula adherens. We also shed light on the endosomal sorting of E-cadherin by showing how Rab7 and Snx16 cooperate in moving E-cadherin into the Rab11 compartment. Thus, our data help to understand how polarised E-cadherin secretion maintains epithelial architecture and prevents metastasis.


Subject(s)
Cadherins/metabolism , Myosin Type V/metabolism , Actins/metabolism , Adherens Junctions/metabolism , Animals , Cell Adhesion , Endosomes/metabolism , Exocytosis , Humans , Neoplasm Metastasis/prevention & control
8.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Article in English | MEDLINE | ID: mdl-34607951

ABSTRACT

Cancer therapy reduces tumor burden via tumor cell death ("debris"), which can accelerate tumor progression via the failure of inflammation resolution. Thus, there is an urgent need to develop treatment modalities that stimulate the clearance or resolution of inflammation-associated debris. Here, we demonstrate that chemotherapy-generated debris stimulates metastasis by up-regulating soluble epoxide hydrolase (sEH) and the prostaglandin E2 receptor 4 (EP4). Therapy-induced tumor cell debris triggers a storm of proinflammatory and proangiogenic eicosanoid-driven cytokines. Thus, targeting a single eicosanoid or cytokine is unlikely to prevent chemotherapy-induced metastasis. Pharmacological abrogation of both sEH and EP4 eicosanoid pathways prevents hepato-pancreatic tumor growth and liver metastasis by promoting macrophage phagocytosis of debris and counterregulating a protumorigenic eicosanoid and cytokine storm. Therefore, stimulating the clearance of tumor cell debris via combined sEH and EP4 inhibition is an approach to prevent debris-stimulated metastasis and tumor growth.


Subject(s)
Eicosanoids/metabolism , Epoxide Hydrolases/biosynthesis , Macrophages/immunology , Neoplasm Metastasis/pathology , Receptors, Prostaglandin E, EP4 Subtype/biosynthesis , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/pathology , Cell Death/drug effects , Cell Line, Tumor , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/prevention & control , Cytokines/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/prevention & control , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Phagocytosis/immunology , RAW 264.7 Cells
9.
Semin Cancer Biol ; 79: 68-82, 2022 02.
Article in English | MEDLINE | ID: mdl-32201368

ABSTRACT

In the last decade, a large amount of research has focused on elucidating the mechanisms that account for homing disseminated cancer cells (DCCs) from solid tumours to distant organs, which successively progress to overt metastatic disease; this is currently incurable. A better understanding of DCC behaviour is expected to allow detectable metastasis prevention by more effectively targeting 'metastatic seeds before they sprout'. As DCC biology co-evolved with that of the primary tumour, and due to the many similarities between them, the term 'niche' has been borrowed from normal adult stem cells (ASCs) to define the site of DCC metastatic colonisation. Moreover, heterogeneity, survival, protection, stemness and plasticity as well as the prolonged G0-G1 dormant state in the metastatic niche have been the main aspects of intense investigation. Consistent with these findings, in solid cancers with minimal residual disease (MRD), it has been proposed to prolong adjuvant therapy by targeting specific molecular pathway(s) involving DCC dormancy. However, so far, few disappointing clinical data have been reported. As an alternative strategy, because immune-surveillance contributes to the steady state of the DCC population and likely to the G0-G1 state of cancer cells, we have used prolonged immune-modulatory cytostatic chemotherapy, active immune stimulation with an INF-ß/IL-2 sequence or drugs inhibiting myeloid-derived suppressor cell (MDSC)/Treg-mediated immune suppression. This strategy, mainly aimed at boosting the immune response, is based on recent findings suggesting the downregulation of immune escape mechanisms as well as other principal hallmarks during the G0-G1 state and/or in MRD. Preliminary clinical and/or laboratory data suggest the efficacy of this strategy in gastrointestinal and some endocrine-dependent cancers. Following this, we propose therapeutic schedules to prevent DCC activation and proliferation in solid cancers at a high risk of relapse or as maintenance therapy in metastatic patients after complete response (CR) to conventional treatment.


Subject(s)
Immunologic Factors/therapeutic use , Immunotherapy/methods , Neoplasm Metastasis/prevention & control , Neoplasm Recurrence, Local/prevention & control , Neoplasm, Residual/therapy , Neoplastic Cells, Circulating/pathology , Cell Proliferation/drug effects , Humans , Interleukin-2/metabolism , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Neoplasm Metastasis/drug therapy , Neoplasm Recurrence, Local/drug therapy , Neoplasm, Residual/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tumor Escape/drug effects , Tumor Escape/immunology
10.
Cancer Metastasis Rev ; 41(2): 383-404, 2022 06.
Article in English | MEDLINE | ID: mdl-35366154

ABSTRACT

Cancer, especially when it has metastasized to different locations in the body, is notoriously difficult to treat. Metastatic cancer accounts for most cancer deaths and thus remains an enormous challenge. During the metastasis process, cancer cells negotiate a series of steps termed the "metastatic cascadeË® that offer potential for developing anti-metastatic therapy strategies. Currently available conventional treatment and diagnostic methods addressing metastasis come with their own pitfalls and roadblocks. In this contribution, we comprehensively discuss the potential improvements that nanotechnology-aided approaches are able to bring, either alone or in combination with the existing conventional techniques, to the identification and treatment of metastatic disease. We tie specific nanotechnology-aided strategies to the complex biology of the different steps of the metastatic cascade in order to open up new avenues for fine-tuned targeting and development of anti-metastatic agents designed specifically to prevent or mitigate the metastatic outgrowth of cancer. We also present a viewpoint on the progress of translation of nanotechnology into cancer metastasis patient care.


Subject(s)
Nanotechnology , Neoplasm Metastasis , Humans , Nanotechnology/methods , Neoplasm Metastasis/prevention & control , Neoplasms/pathology
11.
Nature ; 546(7659): 498-503, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28607485

ABSTRACT

The mutant form of the GTPase KRAS is a key driver of pancreatic cancer but remains a challenging therapeutic target. Exosomes are extracellular vesicles generated by all cells, and are naturally present in the blood. Here we show that enhanced retention of exosomes, compared to liposomes, in the circulation of mice is likely due to CD47-mediated protection of exosomes from phagocytosis by monocytes and macrophages. Exosomes derived from normal fibroblast-like mesenchymal cells were engineered to carry short interfering RNA or short hairpin RNA specific to oncogenic KrasG12D, a common mutation in pancreatic cancer. Compared to liposomes, the engineered exosomes (known as iExosomes) target oncogenic KRAS with an enhanced efficacy that is dependent on CD47, and is facilitated by macropinocytosis. Treatment with iExosomes suppressed cancer in multiple mouse models of pancreatic cancer and significantly increased overall survival. Our results demonstrate an approach for direct and specific targeting of oncogenic KRAS in tumours using iExosomes.


Subject(s)
Exosomes/metabolism , Gene Silencing , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Animals , CD47 Antigen/metabolism , Disease Models, Animal , Exosomes/immunology , Female , Genetic Therapy , Liposomes/immunology , Mice , Monocytes/cytology , Monocytes/immunology , Neoplasm Metastasis/prevention & control , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/pathology , Pinocytosis , Proto-Oncogene Proteins p21(ras)/metabolism , Survival Rate
12.
Nature ; 541(7635): 41-45, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27974793

ABSTRACT

The fact that the identity of the cells that initiate metastasis in most human cancers is unknown hampers the development of antimetastatic therapies. Here we describe a subpopulation of CD44bright cells in human oral carcinomas that do not overexpress mesenchymal genes, are slow-cycling, express high levels of the fatty acid receptor CD36 and lipid metabolism genes, and are unique in their ability to initiate metastasis. Palmitic acid or a high-fat diet specifically boosts the metastatic potential of CD36+ metastasis-initiating cells in a CD36-dependent manner. The use of neutralizing antibodies to block CD36 causes almost complete inhibition of metastasis in immunodeficient or immunocompetent orthotopic mouse models of human oral cancer, with no side effects. Clinically, the presence of CD36+ metastasis-initiating cells correlates with a poor prognosis for numerous types of carcinomas, and inhibition of CD36 also impairs metastasis, at least in human melanoma- and breast cancer-derived tumours. Together, our results indicate that metastasis-initiating cells particularly rely on dietary lipids to promote metastasis.


Subject(s)
Antibodies, Neutralizing/pharmacology , CD36 Antigens/antagonists & inhibitors , Mouth Neoplasms/pathology , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , CD36 Antigens/genetics , CD36 Antigens/immunology , CD36 Antigens/metabolism , Cell Proliferation , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/metabolism , Lipid Metabolism/genetics , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Male , Mice , Mouth Neoplasms/diagnosis , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Neoplastic Stem Cells/metabolism , Palmitic Acid/administration & dosage , Palmitic Acid/metabolism , Palmitic Acid/pharmacology , Penetrance , Prognosis , Transcriptome , Xenograft Model Antitumor Assays
13.
Biol Pharm Bull ; 46(6): 773-780, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37062728

ABSTRACT

This study aimed to determine the effect of brazilin on the invasion and metastasis of breast cancer. The breast cancer MDA-MB-231 and 4T1 cells were treated with brazilin to investigate proliferation and invasion using cell proliferation assay, wound healing assay, transwell assay. BALB/C mice were randomized into normal, model, positive control, and Sappan L. extract groups (n = 6/group). The mice were injected with 4T1 cells via caudal veins to establish a lung metastasis model and via subcutaneous injection to establish a xenograft model. Metastatic nodules on the lung surface, survival rates and visceral indices were evaluated. Subcutaneous tumor volumes and weights were measured. Brazilin inhibited the proliferation of breast cancer cells and significantly inhibited the wound healing, migration, and invasion of MDA-MB-231 and 4T1 cells. Compared with the normal group, the average survival days and spleen index in the model group were significantly decreased, but the lung index and number of pulmonary metastatic nodules were significantly increased. Compared with the model group, the average survival and spleen index of dose groups were significantly increased, and the lung index, the number of pulmonary metastatic nodules, and tumor volume and weight were significantly decreased. Brazilin significantly inhibits the proliferation and metastasis of breast cancer. This study might suggest a new therapeutic agent for breast cancer.


Subject(s)
Breast Neoplasms , Lung Neoplasms , Humans , Animals , Mice , Female , Cell Line, Tumor , Mice, Inbred BALB C , Breast Neoplasms/metabolism , Benzopyrans/pharmacology , Lung Neoplasms/pathology , Cell Movement , Cell Proliferation , Neoplasm Metastasis/prevention & control
14.
Prostate ; 82(2): 245-253, 2022 02.
Article in English | MEDLINE | ID: mdl-34762317

ABSTRACT

BACKGROUND: Patients with high-risk prostate cancer (PC) can experience biochemical relapse (BCR), despite surgery, and develop noncurative disease. The present study aimed to reduce the risk of BCR with a personalized dendritic cell (DC) vaccine, given as adjuvant therapy, after robot-assisted laparoscopic prostatectomy (RALP). METHODS: Twelve weeks after RALP, 20 patients with high-risk PC and undetectable PSA received DC vaccinations for 3 years or until BCR. The primary endpoint was the time to BCR. The immune response was assessed 7 weeks after surgery (baseline) and at one-time point during the vaccination period. RESULTS: Among 20 patients, 11 were BCR-free over a median of 96 months (range: 84-99). The median time from the end of vaccinations to the last follow-up was 57 months (range: 45-60). Nine patients developed BCR, either during (n = 4) or after (n = 5) the vaccination period. Among five patients diagnosed with intraductal carcinoma, three experienced early BCR during the vaccination period. All patients that developed BCR remained in stable disease within a median of 99 months (range: 74-99). The baseline immune response was significantly associated with the immune response during the vaccination period (p = 0.015). For patients diagnosed with extraprostatic extension (EPE), time to BCR was longer in vaccine responders than in non-responders (p = 0.09). Among 12 patients with the International Society of Urological Pathology (ISUP) grade 5 PC, five achieved remission after 84 months, and all mounted immune responses. CONCLUSION: Patients diagnosed with EPE and ISUP grade 5 PC were at particularly high risk of developing postsurgical BCR. In this subgroup, the vaccine response was related to a reduced BCR incidence. The vaccine was safe, without side effects. This adjuvant first-in-man Phase I/II DC vaccine study showed promising results. DC vaccines after curative surgery should be investigated further in a larger cohort of patients with high-risk PC.


Subject(s)
Cancer Vaccines/administration & dosage , Neoplasm Metastasis/prevention & control , Prostate , Prostatectomy/adverse effects , Prostatic Neoplasms , Secondary Prevention/methods , Biomarkers/blood , Dendritic Cells/immunology , Humans , Male , Middle Aged , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Prostate/immunology , Prostate/pathology , Prostate-Specific Antigen/blood , Prostatectomy/methods , Prostatic Neoplasms/blood , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Survival Analysis , Time , Vaccines, Synthetic/administration & dosage
15.
Nature ; 537(7620): 422-426, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27580028

ABSTRACT

Melanoma is the deadliest form of commonly encountered skin cancer because of its rapid progression towards metastasis. Although metabolic reprogramming is tightly associated with tumour progression, the effect of metabolic regulatory circuits on metastatic processes is poorly understood. PGC1α is a transcriptional coactivator that promotes mitochondrial biogenesis, protects against oxidative stress and reprograms melanoma metabolism to influence drug sensitivity and survival. Here, we provide data indicating that PGC1α suppresses melanoma metastasis, acting through a pathway distinct from that of its bioenergetic functions. Elevated PGC1α expression inversely correlates with vertical growth in human melanoma specimens. PGC1α silencing makes poorly metastatic melanoma cells highly invasive and, conversely, PGC1α reconstitution suppresses metastasis. Within populations of melanoma cells, there is a marked heterogeneity in PGC1α levels, which predicts their inherent high or low metastatic capacity. Mechanistically, PGC1α directly increases transcription of ID2, which in turn binds to and inactivates the transcription factor TCF4. Inactive TCF4 causes downregulation of metastasis-related genes, including integrins that are known to influence invasion and metastasis. Inhibition of BRAFV600E using vemurafenib, independently of its cytostatic effects, suppresses metastasis by acting on the PGC1α-ID2-TCF4-integrin axis. Together, our findings reveal that PGC1α maintains mitochondrial energetic metabolism and suppresses metastasis through direct regulation of parallel acting transcriptional programs. Consequently, components of these circuits define new therapeutic opportunities that may help to curb melanoma metastasis.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma/genetics , Melanoma/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/prevention & control , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Transcription, Genetic , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Energy Metabolism , Humans , Indoles/pharmacology , Indoles/therapeutic use , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Integrins/genetics , Integrins/metabolism , Male , Mice , Mitochondria/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/drug therapy , Organelle Biogenesis , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/deficiency , Signal Transduction/drug effects , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Transcription Factor 4 , Transcription Factors/metabolism , Vemurafenib
16.
Acta Pharmacol Sin ; 43(7): 1816-1828, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34785782

ABSTRACT

Omeprazole is a proton pump inhibitor that has recently been reported to exhibit anticancer activity against several types of cancer. However, the anticancer mechanisms of omeprazole remain elusive. Snail is an oncogenic zinc finger transcription factor; aberrant activation of Snail is associated with the occurrence and progression of cancer. In this study, we investigated whether Snail acted as a direct anticancer target of omeprazole. We showed that omeprazole displayed a high binding-affinity to recombinant Snail protein (Kd = 0.076 mM), suggesting that omeprazole directly and physically binds to the Snail protein. We further revealed that omeprazole disrupted CREB-binding protein (CBP)/p300-mediated Snail acetylation and then promoted Snail degradation through the ubiquitin-proteasome pathway in HCT116 cells. Omeprazole treatment markedly suppressed Snail-driven epithelial-mesenchymal transition (EMT) in aggressive HCT116, SUM159, and 4T1 cancer cells in vitro and reduced EMT-associated tumor invasion and metastasis in cancer cell xenograft models. Omeprazole also inhibited tumor growth by limiting Snail-dependent cell cycle progression. Overall, this study, for the first time, identifies Snail as a target of omeprazole and reveals a novel mechanism underlying the therapeutic effects of omeprazole against cancer. This study strongly suggests that omeprazole may be an excellent auxiliary drug for treating patients with malignant tumors.


Subject(s)
Epithelial-Mesenchymal Transition , Omeprazole , Animals , Cell Line, Tumor , Cell Movement , Humans , Mice , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/prevention & control , Omeprazole/pharmacology , Omeprazole/therapeutic use , Snail Family Transcription Factors/metabolism , Transcription Factors/metabolism
17.
Acta Pharmacol Sin ; 43(4): 1001-1012, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34183757

ABSTRACT

Breast cancer is the second leading cause of cancer-related mortality in women, mainly due to metastasis, which is strongly associated with cancer stemness. Our previous studies showed that the eradication of cancer stem-like cells (CSCs) may be related to the activation of dopamine D1 receptor (D1DR). This study aimed to explicitly demonstrate the target-role of D1DR activation in antimetastatic therapy and to investigate the potential efficacy and the underlying D1DR-related mechanisms of QAP14, a new oral compound. 4T1, MDA-MB-231, and D1DR-knockout 4T1 (4T1-D1DR) cells were selected for in vitro study, while 4T1 and 4T1-D1DR cells were further used to establish a mouse allograft model for in vivo study. Our results showed that D1DR is abundantly expressed in both 4T1 and MDA-MB-231 cells and that knocking out D1DR in 4T1 cells accelerated migration and invasion in vitro as well as lung metastasis in vivo. QAP14 inhibited colony formation, cell motility, mammosphere formation and CSC frequency, induced CSC apoptosis and D1DR expression, and increased cAMP/cGMP levels. Additionally, QAP14 showed inhibitory effects on tumor growth and lung metastasis with acceptable safety in vivo. Knocking out D1DR almost completely abolished the efficacy, confirming that QAP14 exhibits its anti-CSC and antimetastatic effects through D1DR activation. The underlying mechanisms involved suppression of the nuclear factor κB (NF-κB)/protein kinase B (Akt) pathway and consequent downregulation of both epithelial-to-mesenchymal transition (EMT) process and cancer stemness. In summary, our findings suggest a potential candidate compound, QAP14, as well as a potential target, D1DR, for metastatic breast cancer therapy.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Female , Humans , Mice , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Neoplastic Stem Cells , Receptors, Dopamine D1/metabolism
18.
Acta Pharmacol Sin ; 43(7): 1829-1842, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34795411

ABSTRACT

Hydrogen sulfide (H2S) is widely recognized as the third endogenous gas signaling molecule and may play a key role in cancer biological processes. ADT-OH (5-(4-hydroxyphenyl)-3H-1,2-dithiocyclopentene-3-thione) is one of the most widely used organic donors for the slow release of H2S and considered to be a potential anticancer compound. In this study, we investigated the antimetastatic effects of ADT-OH in highly metastatic melanoma cells. A tail-vein-metastasis model was established by injecting B16F10 and A375 cells into the tail veins of mice, whereas a mouse footpad-injection model was established by injecting B16F10 cells into mouse footpads. We showed that administration of ADT-OH significantly inhibited the migration and invasion of melanoma cells in the three different animal models. We further showed that ADT-OH dose-dependently inhibited the migration and invasion of B16F10, B16F1 and A375 melanoma cells as evaluated by wound healing and Transwell assays in vitro. LC-MS/MS and bioinformatics analyses revealed that ADT-OH treatment inhibited the EMT process in B16F10 and A375 cells by reducing the expression of FAK and the downstream response protein Paxillin. Overexpression of FAK reversed the inhibitory effects of ADT-OH on melanoma cell migration. Moreover, after ADT-OH treatment, melanoma cells showed abnormal expression of the H2S-producing enzymes CSE/CBS and the AKT signaling pathways. In addition, ADT-OH significantly suppressed the proliferation of melanoma cells. Collectively, these results demonstrate that ADT-OH inhibits the EMT process in melanoma cells by suppressing the CSE/CBS and FAK signaling pathways, thereby exerting its antimetastatic activity. ADT-OH may be used as an antimetastatic agent in the future.


Subject(s)
Melanoma , Thiones , Animals , Cell Line, Tumor , Cell Movement , Chromatography, Liquid , Focal Adhesion Kinase 1/metabolism , Melanoma/drug therapy , Mice , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/prevention & control , Paxillin , Signal Transduction , Skin Neoplasms , Tandem Mass Spectrometry , Melanoma, Cutaneous Malignant
19.
Biochem J ; 478(3): 597-617, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33600595

ABSTRACT

A sequence of interconnected events known as the metastatic cascade promotes tumor progression by regulating cellular and molecular interactions between tumor, stromal, endothelial, and immune cells both locally and systemically. Recently, a new concept has emerged to better describe this process by defining four attributes that metastatic cells should undergo. Every individual hallmark represents a unique trait of a metastatic cell that impacts directly in the outcome of the metastasis process. These critical features, known as the hallmarks of metastasis, include motility and invasion, modulation of the microenvironment, cell plasticity and colonization. They are hierarchically regulated at different levels by several factors, including galectins, a highly conserved family of ß-galactoside-binding proteins abundantly expressed in tumor microenvironments and sites of metastasis. In this review, we discuss the role of galectins in modulating each hallmark of metastasis, highlighting novel therapeutic opportunities for treating the metastatic disease.


Subject(s)
Galectins/physiology , Neoplasm Metastasis/prevention & control , Neoplasm Proteins/physiology , Adaptive Immunity , Animals , Antibodies, Neutralizing/pharmacology , Aptamers, Nucleotide/pharmacology , Carbohydrates/pharmacology , Cell Movement , Clinical Trials, Phase I as Topic , Epithelial-Mesenchymal Transition/physiology , Extracellular Matrix/metabolism , Galectins/antagonists & inhibitors , Humans , Immunity, Innate , Mice , Neoplasm Invasiveness , Neoplasm Metastasis/immunology , Neoplasm Metastasis/physiopathology , Neoplasm Proteins/antagonists & inhibitors , Neoplastic Cells, Circulating , Neovascularization, Pathologic/metabolism , Oligopeptides/pharmacology , Peptides/pharmacology , Polysaccharides/physiology , RNA, Small Interfering/pharmacology , Stromal Cells/metabolism , Tumor Microenvironment/physiology
20.
Proc Natl Acad Sci U S A ; 116(43): 21704-21714, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31591235

ABSTRACT

Metastatic behavior varies significantly among breast cancers. Mechanisms explaining why the majority of breast cancer patients never develop metastatic outgrowth are largely lacking but could underlie the development of novel immunotherapeutic target molecules. Here we show interplay between nonmetastatic primary breast cancer and innate immune response, acting together to control metastatic progression. The primary tumor systemically recruits IFNγ-producing immune effector monocytes to the lung. IFNγ up-regulates Tmem173/STING in neutrophils and enhances their killing capacity. The immune effector monocytes and tumoricidal neutrophils target disseminated tumor cells in the lungs, preventing metastatic outgrowth. Importantly, our findings could underlie the development of immunotherapeutic target molecules that augment the function of immune effector monocytes and neutrophils.


Subject(s)
Cytotoxicity, Immunologic/immunology , Mammary Neoplasms, Animal/pathology , Monocytes/immunology , Neutrophils/immunology , Animals , Cell Line, Tumor , Female , Immunity, Innate/immunology , Immunotherapy/methods , Interferon-gamma/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis/immunology , Neoplasm Metastasis/prevention & control , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL