Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Immunol Immunother ; 65(1): 13-24, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26559812

RESUMO

While inflammation has been associated with the development and progression of colorectal cancer, the exact role of the inflammatory Th17 pathway remains unclear. In this study, we aimed to determine the relative importance of IL-17A and the master regulator of the Th17 pathway, the transcription factor RORγt, in the sporadic intestinal neoplasia of APC(MIN/+) mice and in human colorectal cancer. We show that levels of IL-17A are increased in human colon cancer as compared to adjacent uninvolved colon. Similarly, naïve helper T cells from colorectal cancer patients are more inducible into the Th17 pathway. Furthermore, IL-17A, IL-21, IL-22, and IL-23 are all demonstrated to be directly mitogenic to human colorectal cancer cell lines. Nevertheless, deficiency of IL-17A but not RORγt is associated with decreased spontaneous intestinal tumorigenesis in the APC(MIN/+) mouse model, despite the fact that helper T cells from RORγt-deficient APC(MIN/+) mice do not secrete IL-17A when subjected to Th17-polarizing conditions and that Il17a expression is decreased in the intestine of RORγt-deficient APC(MIN/+) mice. Differential expression of Th17-associated cytokines between IL-17A-deficient and RORγt-deficient APC(MIN/+) mice may explain the difference in adenoma development.


Assuntos
Interleucina-17/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Carcinogênese , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica , Feminino , Humanos , Masculino , Camundongos
2.
Br J Haematol ; 169(1): 36-43, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25572917

RESUMO

Once-weekly administration of bortezomib has reduced bortezomib-induced peripheral neuropathy without affecting response rates, but this has only been demonstrated prospectively in three- and four- drug combinations. We report a phase II trial of alternate dosing and schedule of bortezomib and dexamethasone in newly diagnosed multiple myeloma patients who are not eligible for or refused autologous stem cell transplantation. Bortezomib 1·6 mg/m(2) intravenously was given once-weekly for six cycles, together with dexamethasone 40 mg on the day of and day after bortezomib. Fifty patients were enrolled; 58% did not require any dose modification. The majority of patients had multiple co-morbidities, including cardiovascular (76%) and renal insufficiency (54%), and the median number of medications prior to enrollment was 13. Of all evaluable patients, the overall response rate was 79% and at least 45% had at least a very good partial response. The median time to first response was 1·3 months (range, 0·25-2·4 months). The progression-free and overall survivals were 8 months and 46·5 months, respectively. Twenty-four percent developed worsening neuropathy. We conclude that alternate dosing and scheduling of bortezomib and dexamethasone is both safe and effective for management of newly diagnosed multiple myeloma in frail patients. (ClinicalTrials.gov number, NCT01090921).


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Mieloma Múltiplo , Veteranos , Idoso , Idoso de 80 Anos ou mais , Autoenxertos , Ácidos Borônicos/administração & dosagem , Bortezomib , Dexametasona , Intervalo Livre de Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/mortalidade , Pirazinas/administração & dosagem , Transplante de Células-Tronco , Taxa de Sobrevida
3.
Blood Adv ; 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38861273

RESUMO

Venetoclax is the first example of personalized medicine for multiple myeloma (MM), with meaningful clinical activity as a monotherapy and in combination in myeloma patients harboring the t(11:14) translocation. However, despite the high response rates and prolonged PFS, a significant proportion of patients eventually relapse. Here, we aimed to study adaptive molecular responses after the acquisition of venetoclax resistance in sensitive t(11:14) MM cell models. We therefore generated single-cell venetoclax-resistant t(11:14) MM cell lines and investigated the mechanisms contributing to resistance as well as the cells' sensitivity to other treatments. Our data suggests that acquired resistance to venetoclax is characterized by reduced mitochondrial priming and changes in BCL-2 family proteins' expression in MM cells, conferring broad resistance to standard-of-care anti-myeloma drugs. However, our results show that the resistant cells are still sensitive to immunotherapeutic treatments, highlighting the need to consider appropriate sequencing of these treatments following venetoclax-based regimens.

4.
Blood ; 115(14): 2827-34, 2010 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-19965618

RESUMO

Multiple myeloma (MM) is a plasma cell neoplasm that proceeds through a premalignant state of monoclonal gammopathy of unknown significance; however, the molecular events responsible for myelomagenesis remain uncharacterized. To identify cellular pathways deregulated in MM, we addressed that sumoylation is homologous to ubiquitination and results in the attachment of the ubiquitin-like protein Sumo onto target proteins. Sumoylation was markedly enhanced in MM patient lysates compared with normal plasma cells and expression profiling indicated a relative induction of sumoylation pathway genes. The Sumo-conjugating enzyme Ube2I, the Sumo-ligase PIAS1, and the Sumo-inducer ARF were elevated in MM patient samples and cell lines. Survival correlated with expression because 80% of patients with low UBE2I and PIAS1 were living 6 years after transplantation, whereas only 45% of patients with high expression survived 6 years. UBE2I encodes the sole Sumo-conjugating enzyme in mammalian cells and cells transfected with a dominant-negative sumoylation-deficient UBE2I mutant exhibited decreased survival after radiation exposure, impaired adhesion to bone marrow stroma cell and decreased bone marrow stroma cell-induced proliferation. UBE2I confers cells with multiple advantages to promote tumorigenesis and predicts decreased survival when combined with PIAS1. The sumoylation pathway is a novel therapeutic target with implications for existing proteasomal-based treatment strategies.


Assuntos
Mieloma Múltiplo/metabolismo , Plasmócitos/metabolismo , Processamento de Proteína Pós-Traducional , Proteína SUMO-1/metabolismo , Células da Medula Óssea/metabolismo , Adesão Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Masculino , Mieloma Múltiplo/genética , Mieloma Múltiplo/mortalidade , Mieloma Múltiplo/terapia , Mutação , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Inibidoras de STAT Ativados/biossíntese , Proteína SUMO-1/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/biossíntese , Transplante de Células-Tronco , Células Estromais/metabolismo , Transplante Homólogo , Enzimas de Conjugação de Ubiquitina/biossíntese
5.
Blood ; 115(26): 5385-92, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20395418

RESUMO

Elevated cytokines in bone marrow (BM) micro-environment (interleukin-6 [IL-6], transforming growth factor-beta [TGF-beta], and IL-1beta) may play an important role in observed immune dysfunction in multiple myeloma (MM). As IL-6 and TGF-beta are important for the generation of T-helper 17 (T(H)17) cells, we evaluated and observed a significantly elevated baseline and induced frequency of T(h)17 cells in peripheral blood mononuclear cells (PBMCs) and BM mononuclear cells (BMMCs) from MM patients compared with healthy donors. We observed significant increase in levels of serum IL-17, IL-21, IL-22, and IL-23 in blood and BM in MM compared with healthy donors. We also observed that myeloma PBMCs after T(H)17 polarization significantly induced IL-1alpha, IL-13, IL-17, and IL-23 production compared with healthy donor PBMCs. We next observed that IL-17 promotes myeloma cell growth and colony formation via IL-17 receptor, adhesion to bone marrow stromal cells (BMSCs) as well as increased growth in vivo in murine xenograft model of human MM. Additionally, we have observed that combination of IL-17 and IL-22 significantly inhibited the production of T(H)1-mediated cytokines, including interferon-gamma (IFN-gamma), by healthy donor PBMCs. In conclusion, IL-17-producing T(h)17 cells play an important role in MM pathobiology and may be an important therapeutic target for anti-MM activity and to improve immune function.


Assuntos
Proliferação de Células , Interleucina-17/imunologia , Mieloma Múltiplo/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Citocinas/imunologia , Regulação Neoplásica da Expressão Gênica , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/imunologia , Masculino , Camundongos , Camundongos SCID , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/imunologia , Linfócitos T Auxiliares-Indutores/citologia , Células Th1/citologia , Células Th1/imunologia
6.
Lancet Haematol ; 9(2): e143-e161, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35114152

RESUMO

Infection remains the leading cause of morbidity and mortality in patients with multiple myeloma because of the cumulative effect of disease, treatment, and host-related factors. Given that infectious risk is cumulative through the course of the disease, preventing infections is paramount. Optimal preventive strategies include vaccination against common pathogens, antimicrobial prophylaxis, infection control measures, and immunoglobulin replacement in a small subset of patients; however, there are no universally accepted guidelines for infection prevention. This Review provides a consensus statement from a panel of 36 experts with global representation, which was convened by The International Myeloma Society to review existing literature and current guidelines, address issues associated with the risk of infection and prevention of infectious complications in multiple myeloma in the context of emerging therapies, and offer recommendations for preventing these complications.


Assuntos
Infecções , Mieloma Múltiplo , Consenso , Humanos , Infecções/complicações , Mieloma Múltiplo/complicações , Mieloma Múltiplo/tratamento farmacológico
7.
Blood ; 114(15): 3276-84, 2009 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-19587378

RESUMO

The transformation from monoclonal gammopathy of undetermined significance (MGUS) to multiple myeloma (MM) is thought to be associated with changes in immune processes. We have therefore used serologic analysis of recombinant cDNA expression library to screen the sera of MGUS patients to identify tumor-associated antigens. A total of 10 antigens were identified, with specific antibody responses in MGUS. Responses appeared to be directed against intracellular proteins involved in cellular functions, such as apoptosis (SON, IFT57/HIPPI), DNA and RNA binding (ZNF292, GPATCH4), signal transduction regulators (AKAP11), transcriptional corepressor (IRF2BP2), developmental proteins (OFD1), and proteins of the ubiquitin-proteasome pathway (PSMC1). Importantly, the gene responsible for the oral-facial-digital type I syndrome (OFD1) had response in 6 of 29 (20.6%) MGUS patients but 0 of 11 newly diagnosed MM patients. Interestingly, 3 of 11 (27.2%) MM patients after autologous stem cell transplantations showed responses to OFD1. We have confirmed T-cell responses against OFD1 in MGUS and observed down-regulation of GLI1/PTCH1 and p-beta-catenin after OFD1 knock-down with specific siRNA, suggesting its functional role in the regulation of Hh and Wnt pathways. These findings demonstrate OFD1 as an important immune target and highlight its possible role in signal transduction and tumorigenesis in MGUS and MM.


Assuntos
Autoantígenos/imunologia , Mieloma Múltiplo/imunologia , Paraproteinemias/imunologia , Proteínas/imunologia , Idoso , Idoso de 80 Anos ou mais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Autoantígenos/genética , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , DNA Complementar/genética , Proteínas de Ligação a DNA , Feminino , Humanos , Masculino , Mieloma Múltiplo/genética , Mieloma Múltiplo/terapia , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Paraproteinemias/genética , Paraproteinemias/terapia , Receptores Patched , Receptor Patched-1 , Proteínas/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Transdução de Sinais/imunologia , Transplante de Células-Tronco , Linfócitos T/imunologia , Fatores de Transcrição , Transplante Autólogo , beta Catenina/genética , beta Catenina/imunologia
8.
Proc Natl Acad Sci U S A ; 105(4): 1285-90, 2008 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-18202175

RESUMO

Monoclonal gammopathy of undetermined significance (MGUS) is a common disorder of aging and a precursor lesion to full-blown multiple myeloma (MM). The mechanisms underlying the progression from MGUS to MM are incompletely understood but include the suppression of innate and adaptive antitumor immunity. Here, we demonstrate that NKG2D, an activating receptor on natural killer (NK) cells, CD8(+) T lymphocytes, and MHC class I chain-related protein A (MICA), an NKG2D ligand induced in malignant plasma cells through DNA damage, contribute to the pathogenesis of MGUS and MM. MICA expression is increased on plasma cells from MGUS patients compared with normal donors, whereas MM patients display intermediate MICA levels and a high expression of ERp5, a protein disulfide isomerase linked to MICA shedding (sMICA). MM, but not MGUS, patients harbor circulating sMICA, which triggers the down-regulation of NKG2D and impaired lymphocyte cytotoxicity. In contrast, MGUS, but not MM, patients generate high-titer anti-MICA antibodies that antagonize the suppressive effects of sMICA and stimulate dendritic cell cross-presentation of malignant plasma cells. Bortezomib, a proteasome inhibitor with anti-MM clinical efficacy, activates the DNA damage response to augment MICA expression in some MM cells, thereby enhancing their opsonization by anti-MICA antibodies. Together, these findings reveal that the alterations in the NKG2D pathway are associated with the progression from MGUS to MM and raise the possibility that anti-MICA monoclonal antibodies might prove therapeutic for these disorders.


Assuntos
Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Isoanticorpos/metabolismo , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Ácidos Borônicos/farmacologia , Ácidos Borônicos/uso terapêutico , Bortezomib , Linhagem Celular Tumoral , Apresentação Cruzada , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Progressão da Doença , Antígenos de Histocompatibilidade Classe I/biossíntese , Humanos , Isoanticorpos/uso terapêutico , Ligantes , Mieloma Múltiplo/metabolismo , Prognóstico , Pirazinas/farmacologia , Pirazinas/uso terapêutico , Células Tumorais Cultivadas
9.
Oncotarget ; 12(24): 2323-2337, 2021 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-34853656

RESUMO

CCL20-CCR6 interactions promote colorectal cancer through direct effects on neoplastic epithelial cells and through modulating the tumor microenvironment. The mechanism of these effects on neoplastic epithelial cells is poorly understood. This study demonstrates that CCL20 induces secretion of hepatocyte growth factor (HGF) and phosphorylation of HGF's cognate receptor c-Met in HT29 and HCT116 colorectal cancer cell lines both in concentration- and time-dependent manners. Similar to CCL20, HGF induces migration, autofeedback CCL20 secretion, and ERK1/2 phosphorylation in the colon cancer cells. CCL20-dependent ERK1/2 phosphorylation is blocked by HGF inhibition, and CCL20-dependent migration and CCL20 secretion are blocked by inhibition of HGF or ERK. Interestingly, unlike CCL20, HGF does not induce proliferation of colon cancer cells, and CCL20-dependent cell proliferation is not blocked by direct HGF inhibition. CCL20-dependent proliferation, however, is blocked by the multi-tyrosine kinase inhibitor crizotinib. Exploring this effect, it was found that CCL20 also induces production of MSP and phosphorylation of MSP's receptor MSPR by the colorectal cancer cells. CCL20-dependent cell proliferation is inhibited by directly blocking MSP-MSPR interactions. Thus, CCL20-mediated migration and CCL20 secretion are regulated through a pathway involving HGF, c-Met, and ERK, while CCL20-mediated proliferation is instead regulated through MSP and its receptor MSPR.

10.
Blood Cancer J ; 11(10): 166, 2021 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-34625538

RESUMO

Multiple myeloma (MM) is a heterogeneous disease characterized by significant genomic instability. Recently, a causal role for the AID/APOBEC deaminases in inducing somatic mutations in myeloma has been reported. We have identified APOBEC/AID as a prominent mutational signature at diagnosis with further increase at relapse in MM. In this study, we identified upregulation of several members of APOBEC3 family (A3A, A3B, A3C, and A3G) with A3G, as one of the most expressed APOBECs. We investigated the role of APOBEC3G in MM and observed that A3G expression and APOBEC deaminase activity is elevated in myeloma cell lines and patient samples. Loss-of and gain-of function studies demonstrated that APOBEC3G significantly contributes to increase in DNA damage (abasic sites and DNA breaks) in MM cells. Evaluation of the impact on genome stability, using SNP arrays and whole genome sequencing, indicated that elevated APOBEC3G contributes to ongoing acquisition of both the copy number and mutational changes in MM cells over time. Elevated APOBEC3G also contributed to increased homologous recombination activity, a mechanism that can utilize increased DNA breaks to mediate genomic rearrangements in cancer cells. These data identify APOBEC3G as a novel gene impacting genomic evolution and underlying mechanisms in MM.


Assuntos
Desaminase APOBEC-3G/metabolismo , Dano ao DNA , Instabilidade Genômica , Mieloma Múltiplo/enzimologia , Mutação , Proteínas de Neoplasias/metabolismo , Desaminase APOBEC-3G/genética , Linhagem Celular Tumoral , Humanos , Mieloma Múltiplo/genética , Proteínas de Neoplasias/genética
11.
Blood Cancer Discov ; 2(5): 468-483, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34568832

RESUMO

Proteasome inhibitor bortezomib induces apoptosis in multiple myeloma (MM) cells, and has transformed patient outcome. Using in vitro as well as in vivo immunodeficient and immunocompetent murine MM models, we here show that bortezomib also triggers immunogenic cell death (ICD) characterized by exposure of calreticulin on dying MM cells, phagocytosis of tumor cells by dendritic cells, and induction of MM specific immunity. We identify a bortezomib-triggered specific ICD-gene signature associated with better outcome in two independent MM patient cohorts. Importantly, bortezomib stimulates MM cells immunogenicity via activation of cGAS/STING pathway and production of type-I interferons; and STING agonists significantly potentiate bortezomib-induced ICD. Our studies therefore delineate mechanisms whereby bortezomib exerts immunotherapeutic activity, and provide the framework for clinical trials of STING agonists with bortezomib to induce potent tumor-specific immunity and improve patient outcome in MM.


Assuntos
Mieloma Múltiplo , Animais , Bortezomib/farmacologia , Humanos , Imunidade , Proteínas de Membrana/genética , Camundongos , Mieloma Múltiplo/tratamento farmacológico , Nucleotidiltransferases/genética , Transdução de Sinais
12.
Hematol Oncol Clin North Am ; 21(6): 1217-30, x-xi, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17996595

RESUMO

Immune cells with specific functions and abilities are vital to cancer treatment prevention. Although there have been many accomplishments made in the areas of immunotherapy and immunobiology of myeloma, there are still many obstacles in the way of conceptualizing the interrelationships between immune cells and tumor cells. To provide better understanding of these concepts and to move toward improved therapies for myeloma, cell-based therapeutic approaches should be developed.


Assuntos
Células Dendríticas , Imunoterapia Ativa/métodos , Mieloma Múltiplo/imunologia , Comunicação Celular/imunologia , Ensaios Clínicos como Assunto , Células Dendríticas/imunologia , Células Dendríticas/transplante , Humanos , Idiótipos de Imunoglobulinas/imunologia , Mieloma Múltiplo/tratamento farmacológico
13.
Oncoimmunology ; 6(1): e1261776, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28197386

RESUMO

IL-21 has reported activity in promoting both Th1 and Th17 immune responses. Its role in sporadic human colorectal cancer is unknown. We aimed to delineate the role of IL-21 in a model of sporadic intestinal carcinogenesis. We found that in APCMIN/+ mice, ablation of IL-21 increased intestinal tumorigenesis. Expression of pro-inflammatory Th17-associated genes, including RORγt and IL-17A, was increased in the intestine in the absence of IL-21, while expression of antitumor Th1-associated genes Tbet, IFNγ, granzyme B, and perforin was decreased. Similarly, the IL-21-deficient APCMIN/+ mouse intestines had fewer infiltrating T cells as well as decreased effector memory T cells, NK cells, and granzyme B-expressing cells. Finally, our data suggest that IL-21 impairs Th17 immune responses as mesenteric lymph nodes from IL-21-deficient mice had increased IL-17A expression, and naive helper T cells from IL-21-deficient mice were more prone to differentiate into IL-17A-secreting cells.

14.
Sci Rep ; 7: 45681, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28374831

RESUMO

The necessity for bone marrow aspiration and the lack of highly sensitive assays to detect residual disease present challenges for effective management of multiple myeloma (MM), a plasma cell cancer. We show that a microfluidic cell capture based on CD138 antigen, which is highly expressed on plasma cells, permits quantitation of rare circulating plasma cells (CPCs) in blood and subsequent fluorescence-based assays. The microfluidic device is based on a herringbone channel design, and exhibits an estimated cell capture efficiency of ~40-70%, permitting detection of <10 CPCs/mL using 1-mL sample volumes, which is difficult using existing techniques. In bone marrow samples, the microfluidic-based plasma cell counts exhibited excellent correlation with flow cytometry analysis. In peripheral blood samples, the device detected a baseline of 2-5 CD138+ cells/mL in healthy donor blood, with significantly higher numbers in blood samples of MM patients in remission (20-24 CD138+ cells/mL), and yet higher numbers in MM patients exhibiting disease (45-184 CD138+ cells/mL). Analysis of CPCs isolated using the device was consistent with serum immunoglobulin assays that are commonly used in MM diagnostics. These results indicate the potential of CD138-based microfluidic CPC capture as a useful 'liquid biopsy' that may complement or partially replace bone marrow aspiration.


Assuntos
Anticorpos/metabolismo , Mieloma Múltiplo/patologia , Plasmócitos/citologia , Sindecana-1/metabolismo , Medula Óssea/metabolismo , Medula Óssea/patologia , Linhagem Celular Tumoral , Humanos , Dispositivos Lab-On-A-Chip , Mieloma Múltiplo/metabolismo
15.
Oncoimmunology ; 5(8): e1189052, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27622061

RESUMO

Interactions between the inflammatory chemokine CCL20 and its receptor CCR6 have been implicated in promoting colon cancer; however, the mechanisms behind this effect are poorly understood. We have previously demonstrated that deficiency of CCR6 is associated with decreased tumor macrophage accumulation in a model of sporadic intestinal tumorigenesis. In this study, we aimed to determine the role of stromal CCR6 expression in a murine syngeneic transplantable colon cancer model. We show that deficiency of host CCR6 is associated with decreased growth of syngeneic CCR6-expressing colon cancers. Colon cancers adoptively transplanted into CCR6-deficient mice have decreased tumor-associated macrophages without alterations in the number of monocytes in blood or bone marrow. CCL20, the unique ligand for CCR6, promotes migration of monocytes in vitro and promotes accumulation of macrophages in vivo. Depletion of tumor-associated macrophages decreases the growth of tumors in the transplantable tumor model. Macrophages infiltrating the colon cancers in this model secrete the inflammatory mediators CCL2, IL-1α, IL-6 and TNFα. Ccl2, Il1α and Il6 are consequently downregulated in tumors from CCR6-deficient mice. CCL2, IL-1α and IL-6 also promote proliferation of colon cancer cells, linking the decreased macrophage migration into tumors mediated by CCL20-CCR6 interactions to the delay in tumor growth in CCR6-deficient hosts. The relevance of these findings in human colon cancer is demonstrated through correlation of CCR6 expression with that of the macrophage marker CD163 as well as that of CCL2, IL1α and TNFα. Our findings support the exploration of targeting the CCL20-CCR6 pathway for the treatment of colon cancer.

16.
PLoS One ; 9(5): e97566, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24866282

RESUMO

Interactions between the inflammatory chemokine CCL20 and its receptor CCR6 have been associated with colorectal cancer growth and metastasis, however, a causal role for CCL20 signaling through CCR6 in promoting intestinal carcinogenesis has not been demonstrated in vivo. In this study, we aimed to determine the role of CCL20-CCR6 interactions in spontaneous intestinal tumorigenesis. CCR6-deficient mice were crossed with mice heterozygous for a mutation in the adenomatous polyposis coli (APC) gene (APCMIN/+ mice) to generate APCMIN/+ mice with CCR6 knocked out (CCR6KO-APCMIN/+ mice). CCR6KO-APCMIN/+ mice had diminished spontaneous intestinal tumorigenesis. CCR6KO-APCMIN/+ also had normal sized spleens as compared to the enlarged spleens found in APCMIN/+ mice. Decreased macrophage infiltration into intestinal adenomas and non-tumor epithelium was observed in CCR6KO-APCMIN/+ as compared to APCMIN/+ mice. CCL20 signaling through CCR6 caused increased production of CCL20 by colorectal cancer cell lines. Furthermore, CCL20 had a direct mitogenic effect on colorectal cancer cells. Thus, interactions between CCL20 and CCR6 promote intestinal carcinogenesis. Our results suggest that the intestinal tumorigenesis driven by CCL20-CCR6 interactions may be driven by macrophage recruitment into the intestine as well as proliferation of neoplastic epithelial cells. This interaction could be targeted for the treatment or prevention of malignancy.


Assuntos
Transformação Celular Neoplásica/patologia , Quimiocina CCL20/metabolismo , Neoplasias do Colo/patologia , Genes APC , Neoplasias Intestinais/patologia , Receptores CCR6/metabolismo , Receptores CCR6/fisiologia , Animais , Apoptose , Western Blotting , Proliferação de Células , Transformação Celular Neoplásica/genética , Quimiocina CCL20/genética , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Feminino , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Neoplasias Intestinais/etiologia , Neoplasias Intestinais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores CCR6/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
17.
Blood ; 107(1): 301-4, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16150935

RESUMO

Multiple myeloma (MM) is characterized by the production of monoclonal immunoglobulin and is associated with suppressed uninvolved immunoglobulins and dysfunctional T-cell responses. The biologic basis of this dysfunction remains ill defined. Because T regulatory (T(reg)) cells play an important role in suppressing normal immune responses, we evaluated the potential role of T(reg) cells in immune dysfunction in MM. We observed a significant increase in CD4+ CD25+ T cells in patients with monoclonal gammopathy of undetermined significance (MGUS) and in patients with MM compared with healthy donors (25% and 26%, respectively, vs 14%); however, T(reg) cells as measured by FOXP3 expression are significantly decreased in patients with MGUS and MM compared with healthy donors. Moreover, even when they are added in higher proportions, T(reg) cells in patients with MM and MGUS are unable to suppress anti-CD3-mediated T-cell proliferation. This decreased number and function of T(reg) cells in MGUS and in MM may account, at least in part, for the nonspecific increase in CD4+ CD25+ T cells, thereby contributing to dysfunctional T-cell responses.


Assuntos
Mieloma Múltiplo/imunologia , Linfócitos T Reguladores/patologia , Contagem de Linfócito CD4 , Estudos de Casos e Controles , Proliferação de Células , Humanos , Imunidade Celular , Ativação Linfocitária , Gamopatia Monoclonal de Significância Indeterminada , Linfócitos T Reguladores/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA