Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Aging (Albany NY) ; 13(21): 23981-24016, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34762598

RESUMO

This study addresses the potential to reverse age-associated morbidity by establishing methods to restore the aged hematopoietic system. Parabiotic animal models indicated that young secretome could restore aged tissues, leading us to establish a heterochronic transwell system with aged mobilized peripheral blood (MPB), co-cultured with young MPB or umbilical cord blood (UCB) cells. Functional studies and omics approaches indicate that the miRNA cargo of microvesicles (MVs) restores the aged hematopoietic system. The in vitro findings were validated in immune deficient (NSG) mice carrying an aged hematopoietic system, improving aged hallmarks such as increased lymphoid:myeloid ratio, decreased inflammation and cellular senescence. Elevated MYC and E2F pathways, and decreased p53 were key to hematopoietic restoration. These processes require four restorative miRs that target the genes for transcription/differentiation, namely PAX and phosphatase PPMIF. These miRs when introduced in aged cells were sufficient to restore the aged hematopoietic system in NSG mice. The aged MPBs were the drivers of their own restoration, as evidenced by the changes from distinct baseline miR profiles in MPBs and UCB to comparable expressions after exposure to aged MPBs. Restorative natural killer cells eliminated dormant breast cancer cells in vivo, indicating the broad relevance of this cellular paradigm - preventing and reversing age-associated disorders such as clearance of early malignancies and enhanced responses to vaccine and infection.


Assuntos
Células da Medula Óssea , Micropartículas Derivadas de Células , Senescência Celular/fisiologia , Hematopoese/fisiologia , Adulto , Idoso , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/fisiologia , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/fisiologia , Feminino , Sangue Fetal/citologia , Humanos , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Secretoma , Adulto Jovem
2.
Life Sci Alliance ; 4(7)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34078741

RESUMO

The challenge for treating breast cancer (BC) is partly due to long-term dormancy driven by cancer stem cells (CSCs) capable of evading immune response and resist chemotherapy. BC cells show preference for the BM, resulting in poor prognosis. CSCs use connexin 43 (Cx43) to form gap junctional intercellular communication with BM niche cells, fibroblasts, and mesenchymal stem cells (MSCs). However, Cx43 is an unlikely target to reverse BC dormancy because of its role as a hematopoietic regulator. We found N-cadherin (CDH2) and its associated pathways as potential drug targets. CDH2, highly expressed in CSCs, interacts intracellularly with Cx43, colocalizes with Cx43 in BC cells within BM biopsies of patients, and is required for Cx43-mediated gap junctional intercellular communication with BM niche cells. Notably, CDH2 and anti-apoptotic pathways maintained BC dormancy. We thereby propose these pathways as potential pharmacological targets to prevent dormancy and chemosensitize resistant CSCs.


Assuntos
Antígenos CD/metabolismo , Neoplasias da Mama/metabolismo , Caderinas/metabolismo , Conexina 43/metabolismo , Antígenos CD/genética , Medula Óssea/metabolismo , Caderinas/genética , Caderinas/fisiologia , Conexina 43/genética , Resistencia a Medicamentos Antineoplásicos/fisiologia , Feminino , Junções Comunicantes/metabolismo , Junções Comunicantes/patologia , Humanos , Células-Tronco Mesenquimais/metabolismo , Metástase Neoplásica/patologia , Células-Tronco Neoplásicas/metabolismo , Evasão Tumoral/fisiologia
3.
Methods Mol Biol ; 2224: 195-202, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33606216

RESUMO

Mice with human hematopoietic system have become critical for research and preclinical studies. Mice with patient-derived xenografts of different tumors exist without human immune system. Answers can be addressed with the same immunodeficient mice that are chimeric for the human hemato-lymphoid system (humanized mice). The growing field of immune-oncology could benefit from preclinical studies with the humanized mice. Other fields will also benefit such as studies of infectious disease, regenerative medicine, organ transplant, and allergies. Here, we describe the method to humanize immune-deficient mice with human CD34+ hematopoietic cells.


Assuntos
Sistema Imunitário/imunologia , Síndromes de Imunodeficiência/imunologia , Adulto , Animais , Antígenos CD34/imunologia , Modelos Animais de Doenças , Feminino , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade
4.
J Immunol ; 204(4): 879-891, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31924647

RESUMO

Hematopoiesis is tightly regulated by the bone marrow (BM) niche. The niche is robust, allowing for the return of hematopoietic homeostasis after insults such as infection. Hematopoiesis is partly regulated by soluble factors, such as neuropeptides, substance P (SP), and neurokinin A (NK-A), which mediate hematopoietic stimulation and inhibition, respectively. SP and NK-A are derived from the Tac1 gene that is alternately spliced into four variants. The hematopoietic effects of SP and NK-A are mostly mediated via BM stroma. Array analyses with 2400 genes indicated distinct changes in SP-stimulated BM stroma. Computational analyses indicated networks of genes with hematopoietic regulation. Included among these networks is the high-mobility group box 1 gene (HMGB1), a nonhistone chromatin-associated protein. Validation studies indicated that NK-A could reverse SP-mediated HMGB1 decrease. Long-term culture-initiating cell assay, with or without NK-A receptor antagonist (NK2), showed a suppressive effect of HMGB1 on hematopoietic progenitors and increase in long-term culture-initiating cell assay cells (primitive hematopoietic cells). These effects occurred partly through NK-A. NSG mice with human hematopoietic system injected with the HMGB1 antagonist glycyrrhizin verified the in vitro effects of HMGB1. Although the effects on myeloid lineage were suppressed, the results suggested a more complex effect on the lymphoid lineage. Clonogenic assay for CFU- granulocyte-monocyte suggested that HMGB1 may be required to prevent hematopoietic stem cell exhaustion to ensure immune homeostasis. In summary, this study showed how HMGB1 is linked to SP and NK-A to protect the most primitive hematopoietic cell and also to maintain immune/hematopoietic homeostasis.


Assuntos
Proteína HMGB1/metabolismo , Hematopoese/genética , Neuroimunomodulação/genética , Neurocinina A/metabolismo , Substância P/metabolismo , Adolescente , Adulto , Processamento Alternativo , Animais , Benzamidas/farmacologia , Biópsia , Medula Óssea/metabolismo , Medula Óssea/patologia , Transplante de Medula Óssea , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Redes Reguladoras de Genes/efeitos dos fármacos , Redes Reguladoras de Genes/imunologia , Células HEK293 , Hematopoese/imunologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Neuroimunomodulação/imunologia , Neurocinina A/antagonistas & inibidores , Análise de Sequência com Séries de Oligonucleotídeos , Piperidinas/farmacologia , Cultura Primária de Células , Taquicininas/genética , Quimeras de Transplante , Adulto Jovem
5.
Sci Rep ; 8(1): 367, 2018 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-29321622

RESUMO

This study proposes that a novel developmental hierarchy of breast cancer (BC) cells (BCCs) could predict treatment response and outcome. The continued challenge to treat BC requires stratification of BCCs into distinct subsets. This would provide insights on how BCCs evade treatment and adapt dormancy for decades. We selected three subsets, based on the relative expression of octamer-binding transcription factor 4 A (Oct4A) and then analysed each with Affymetrix gene chip. Oct4A is a stem cell gene and would separate subsets based on maturation. Data analyses and gene validation identified three membrane proteins, TMEM98, GPR64 and FAT4. BCCs from cell lines and blood from BC patients were analysed for these three membrane proteins by flow cytometry, along with known markers of cancer stem cells (CSCs), CD44, CD24 and Oct4, aldehyde dehydrogenase 1 (ALDH1) activity and telomere length. A novel working hierarchy of BCCs was established with the most immature subset as CSCs. This group was further subdivided into long- and short-term CSCs. Analyses of 20 post-treatment blood indicated that circulating CSCs and early BC progenitors may be associated with recurrence or early death. These results suggest that the novel hierarchy may predict treatment response and prognosis.


Assuntos
Biomarcadores Tumorais , Neoplasias da Mama/genética , Biologia Computacional , Perfilação da Expressão Gênica , Transcriptoma , Adulto , Idoso , Idoso de 80 Anos ou mais , Família Aldeído Desidrogenase 1 , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Biologia Computacional/métodos , Feminino , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Imunofenotipagem , Isoenzimas/metabolismo , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Estadiamento de Neoplasias , Retinal Desidrogenase/metabolismo , Homeostase do Telômero
6.
Cancer Res ; 76(19): 5832-5844, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27569215

RESUMO

Dormant breast cancers resurge as metastatic disease after a long dormancy period in the bone marrow, where cancer cells interact with mesenchymal stem cells (MSC). However, the nature of early interactions between breast cancer cells and MSCs in the bone marrow microenvironment that facilitate adaptation to a quiescent state remains poorly understood. Here, we report that breast cancer cells prime MSC to release exosomes containing distinct miRNA contents, such as miR-222/223, which in turn promotes quiescence in a subset of cancer cells and confers drug resistance. Building on these results, we developed a novel, nontoxic therapeutic strategy to target dormant breast cancer cells based on systemic administration of MSC loaded with antagomiR-222/223. In an immunodeficient mouse model of dormant breast cancer, this therapy sensitized breast cancer cells to carboplatin-based therapy and increased host survival. Overall, our findings illuminate the nature of the regulatory interactions between breast cancer cells and MSCs in the evolution of tumor dormancy and resurgence in the micrometastatic microenvironment of the bone marrow. Cancer Res; 76(19); 5832-44. ©2016 AACR.


Assuntos
Medula Óssea/patologia , Neoplasias da Mama/patologia , Exossomos/fisiologia , Células-Tronco Mesenquimais/fisiologia , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/mortalidade , Carboplatina/uso terapêutico , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/antagonistas & inibidores , MicroRNAs/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...