Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Redox Biol ; 70: 103042, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38244399

RESUMO

Hypoxia is the key pathobiological trigger of tubular oxidative stress and cell death that drives the transition of acute kidney injury (AKI) to chronic kidney disease (CKD). The mitochondrial-rich proximal tubular epithelial cells (PTEC) are uniquely sensitive to hypoxia and thus, are pivotal in propagating the sustained tubular loss of AKI-to-CKD transition. Here, we examined the role of PTEC-derived small extracellular vesicles (sEV) in propagating the 'wave of tubular death'. Ex vivo patient-derived PTEC were cultured under normoxia (21 % O2) and hypoxia (1 % O2) on Transwell inserts for isolation and analysis of sEV secreted from apical versus basolateral PTEC surfaces. Increased numbers of sEV were secreted from the apical surface of hypoxic PTEC compared with normoxic PTEC. No differences in basolateral sEV numbers were observed between culture conditions. Biological pathway analysis of hypoxic-apical sEV cargo identified distinct miRNAs linked with cellular injury pathways. In functional assays, hypoxic-apical sEV selectively induced ferroptotic cell death (↓glutathione peroxidase-4, ↑lipid peroxidation) in autologous PTEC compared with normoxic-apical sEV. The addition of ferroptosis inhibitors, ferrostatin-1 and baicalein, attenuated PTEC ferroptosis. RNAse A pretreatment of hypoxic-apical sEV also abrogated PTEC ferroptosis, demonstrating a role for sEV RNA in ferroptotic 'wave of death' signalling. In line with these in vitro findings, in situ immunolabelling of diagnostic kidney biopsies from AKI patients with clinical progression to CKD (AKI-to-CKD transition) showed evidence of ferroptosis propagation (increased numbers of ACSL4+ PTEC), while urine-derived sEV (usEV) from these 'AKI-to-CKD transition' patients triggered PTEC ferroptosis (↑lipid peroxidation) in functional studies. Our data establish PTEC-derived apical sEV and their intravesicular RNA as mediators of tubular lipid peroxidation and ferroptosis in hypoxic kidney injury. This concept of how tubular pathology is propagated from the initiating insult into a 'wave of death' provides novel therapeutic check-points for targeting AKI-to-CKD transition.


Assuntos
Injúria Renal Aguda , Ferroptose , Insuficiência Renal Crônica , Humanos , Túbulos Renais Proximais , Rim/metabolismo , Células Epiteliais/metabolismo , Hipóxia/metabolismo , Injúria Renal Aguda/metabolismo , Insuficiência Renal Crônica/metabolismo , RNA
2.
Cell Death Dis ; 13(8): 739, 2022 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-36030251

RESUMO

Inflammasomes are multiprotein platforms responsible for the release of pro-inflammatory cytokines interleukin (IL)-1ß and IL-18. Mouse studies have identified inflammasome activation within dendritic cells (DC) as pivotal for driving tubulointerstitial fibrosis and inflammation, the hallmarks of chronic kidney disease (CKD). However, translation of this work to human CKD remains limited. Here, we examined the complex tubular cell death pathways mediating inflammasome activation in human kidney DC and, thus, CKD progression. Ex vivo patient-derived proximal tubular epithelial cells (PTEC) cultured under hypoxic (1% O2) conditions modelling the CKD microenvironment showed characteristics of ferroptotic cell death, including mitochondrial dysfunction, reductions in the lipid repair enzyme glutathione peroxidase 4 (GPX4) and increases in lipid peroxidation by-product 4-hydroxynonenal (4-HNE) compared with normoxic PTEC. The addition of ferroptosis inhibitor, ferrostatin-1, significantly reduced hypoxic PTEC death. Human CD1c+ DC activated in the presence of hypoxic PTEC displayed significantly increased production of inflammasome-dependent cytokines IL-1ß and IL-18. Treatment of co-cultures with VX-765 (caspase-1/4 inhibitor) and MCC950 (NLRP3 inflammasome inhibitor) significantly attenuated IL-1ß/IL-18 levels, supporting an NLRP3 inflammasome-dependent DC response. In line with these in vitro findings, in situ immunolabelling of human fibrotic kidney tissue revealed a significant accumulation of tubulointerstitial CD1c+ DC containing active inflammasome (ASC) specks adjacent to ferroptotic PTEC. These data establish ferroptosis as the primary pattern of PTEC necrosis under the hypoxic conditions of CKD. Moreover, this study identifies NLRP3 inflammasome signalling driven by complex tubulointerstitial PTEC-DC interactions as a key checkpoint for therapeutic targeting in human CKD.


Assuntos
Células Dendríticas , Células Epiteliais , Ferroptose , Proteína 3 que Contém Domínio de Pirina da Família NLR , Insuficiência Renal Crônica , Antígenos CD1 , Caspase 1 , Citocinas , Células Dendríticas/citologia , Células Epiteliais/citologia , Fibrose , Glicoproteínas , Humanos , Inflamassomos , Interleucina-18 , Interleucina-1beta , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Insuficiência Renal Crônica/patologia
3.
J Extracell Vesicles ; 10(4): e12064, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33643548

RESUMO

Proximal tubular epithelial cells (PTEC) are central players in inflammatory kidney diseases. However, the complex signalling mechanism/s via which polarized PTEC mediate disease progression are poorly understood. Small extracellular vesicles (sEV), including exosomes, are recognized as fundamental components of cellular communication and signalling courtesy of their molecular cargo (lipids, microRNA, proteins). In this study, we examined the molecular content and function of sEV secreted from the apical versus basolateral surfaces of polarized human primary PTEC under inflammatory diseased conditions. PTEC were cultured under normal and inflammatory conditions on Transwell inserts to enable separate collection and isolation of apical/basolateral sEV. Significantly increased numbers of apical and basolateral sEV were secreted under inflammatory conditions compared with equivalent normal conditions. Multi-omics analysis revealed distinct molecular profiles (lipids, microRNA, proteins) between inflammatory and normal conditions for both apical and basolateral sEV. Biological pathway analyses of significantly differentially expressed molecules associated apical inflammatory sEV with processes of cell survival and immunological disease, while basolateral inflammatory sEV were linked to pathways of immune cell trafficking and cell-to-cell signalling. In line with this mechanistic concept, functional assays demonstrated significantly increased production of chemokines (monocyte chemoattractant protein-1, interleukin-8) and immuno-regulatory cytokine interleukin-10 by peripheral blood mononuclear cells activated with basolateral sEV derived from inflammatory PTEC. We propose that the distinct molecular composition of sEV released from the apical versus basolateral membranes of human inflammatory PTEC may reflect specialized functional roles, with basolateral-derived sEV pivotal in modulating tubulointerstitial inflammatory responses observed in many immune-mediated kidney diseases. These findings provide a rationale to further evaluate these sEV-mediated inflammatory pathways as targets for biomarker and therapeutic development.


Assuntos
Comunicação Celular , Células Epiteliais/metabolismo , Exossomos/fisiologia , Vesículas Extracelulares/fisiologia , Nefropatias/metabolismo , Túbulos Renais Proximais/metabolismo , Transdução de Sinais , Adulto , Transporte Biológico , Biomarcadores , Células Cultivadas , Ceramidas/metabolismo , Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Progressão da Doença , Células Epiteliais/química , Exossomos/química , Vesículas Extracelulares/química , Feminino , Humanos , Inflamação/metabolismo , Leucócitos Mononucleares/metabolismo , Metabolismo dos Lipídeos , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Proteínas/metabolismo , Proteômica
4.
J Am Soc Nephrol ; 30(7): 1322-1335, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31186283

RESUMO

BACKGROUND: Mucosal-associated invariant T (MAIT) cells represent a specialized lymphocyte population associated with chronic inflammatory disorders. Little is known, however, about MAIT cells in diseases of the kidney, including CKD. METHODS: To evaluate MAIT cells in human native kidneys with tubulointerstitial fibrosis, the hallmark of CKD, we used multicolor flow cytometry to identify, enumerate, and phenotype such cells from human kidney tissue biopsy samples, and immunofluorescence microscopy to localize these cells. We cocultured MAIT cells and human primary proximal tubular epithelial cells (PTECs) under hypoxic (1% oxygen) conditions to enable examination of mechanistic tubulointerstitial interactions. RESULTS: We identified MAIT cells (CD3+ TCR Vα7.2+ CD161hi) in healthy and diseased kidney tissues, detecting expression of tissue-resident markers (CD103/CD69) on MAIT cells in both states. Tissue samples from kidneys with tubulointerstitial fibrosis had significantly elevated numbers of MAIT cells compared with either nonfibrotic samples from diseased kidneys or tissue samples from healthy kidneys. Furthermore, CD69 expression levels, also an established marker of lymphocyte activation, were significantly increased on MAIT cells from fibrotic tissue samples. Immunofluorescent analyses of fibrotic kidney tissue identified MAIT cells accumulating adjacent to PTECs. Notably, MAIT cells activated in the presence of human PTECs under hypoxic conditions (modeling the fibrotic microenvironment) displayed significantly upregulated expression of CD69 and cytotoxic molecules perforin and granzyme B; we also observed a corresponding significant increase in PTEC necrosis in these cocultures. CONCLUSIONS: Our findings indicate that human tissue-resident MAIT cells in the kidney may contribute to the fibrotic process of CKD via complex interactions with PTECs.


Assuntos
Rim/patologia , Células T Invariantes Associadas à Mucosa/fisiologia , Insuficiência Renal Crônica/imunologia , Adulto , Idoso , Antígenos CD/análise , Antígenos de Diferenciação de Linfócitos T/análise , Comunicação Celular , Técnicas de Cocultura , Células Epiteliais/fisiologia , Feminino , Fibrose , Humanos , Túbulos Renais Proximais/citologia , Lectinas Tipo C/análise , Masculino , Pessoa de Meia-Idade , Insuficiência Renal Crônica/patologia
5.
Bio Protoc ; 8(16): e2980, 2018 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34395780

RESUMO

Inflammatory immune cells play direct pathological roles in cases of acute kidney injury (AKI) and chronic kidney disease (CKD). However, the identification and characterization of distinct populations of leukocytes in human kidney biopsies have been confounded by the limitations of immunohistochemical (IHC)-based techniques used to detect them. This methodology is not amenable to the combinations of multiple markers necessary to unequivocally define discrete immune cell populations. We have developed a multi-parameter, flow cytometric-based approach that addresses the need for panels of cell-specific markers in the identification of immune cell populations, allowing both the accurate detection and quantitation of leukocyte subpopulations from a single, clinical kidney biopsy specimen. In this approach, fresh human kidney tissue is dissociated into a single cell suspension followed by antibody-labeling and flow cytometric-based acquisition and analysis. This novel technique provides a major step forward in identifying and enumerating immune cell subpopulations in human kidney disease and is a powerful platform to complement traditional histopathological examinations of clinical kidney biopsies.

6.
Kidney Int ; 92(1): 79-88, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28396119

RESUMO

Natural killer (NK) cells are a population of lymphoid cells that play a significant role in mediating innate immune responses. Studies in mice suggest a pathological role for NK cells in models of kidney disease. In this study, we characterized the NK cell subsets present in native kidneys of patients with tubulointerstitial fibrosis, the pathological hallmark of chronic kidney disease. Significantly higher numbers of total NK cells (CD3-CD56+) were detected in renal biopsies with tubulointerstitial fibrosis compared with diseased biopsies without fibrosis and healthy kidney tissue using multi-color flow cytometry. At a subset level, both the CD56dim NK cell subset and particularly the CD56bright NK cell subset were elevated in fibrotic kidney tissue. However, only CD56bright NK cells significantly correlated with the loss of kidney function. Expression of the tissue-retention and -activation molecule CD69 on CD56bright NK cells was significantly increased in fibrotic biopsy specimens compared with non-fibrotic kidney tissue, indicative of a pathogenic phenotype. Further flow cytometric phenotyping revealed selective co-expression of activating receptor CD335 (NKp46) and differentiation marker CD117 (c-kit) on CD56bright NK cells. Multi-color immunofluorescent staining of fibrotic kidney tissue localized the accumulation of NK cells within the tubulointerstitium, with CD56bright NK cells (NKp46+ CD117+) identified as the source of pro-inflammatory cytokine interferon-γ within the NK cell compartment. Thus, activated interferon-γ-producing CD56bright NK cells are positioned to play a key role in the fibrotic process and progression to chronic kidney disease.


Assuntos
Antígeno CD56/análise , Interferon gama/análise , Túbulos Renais/imunologia , Células Matadoras Naturais/imunologia , Insuficiência Renal Crônica/imunologia , Adulto , Idoso , Antígenos CD/análise , Antígenos de Diferenciação de Linfócitos T/análise , Biópsia , Estudos de Casos e Controles , Progressão da Doença , Feminino , Fibrose , Citometria de Fluxo , Imunofluorescência , Humanos , Túbulos Renais/patologia , Células Matadoras Naturais/patologia , Lectinas Tipo C/análise , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Receptor 1 Desencadeador da Citotoxicidade Natural/análise , Proteínas Proto-Oncogênicas c-kit/análise , Insuficiência Renal Crônica/patologia , Transdução de Sinais
7.
Nephrol Dial Transplant ; 30(10): 1674-83, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26058593

RESUMO

BACKGROUND: Descriptions of inflammatory cells infiltrating the human kidney rarely mention B cells, other than in the specific scenario of transplantation. In these reports, B cells are localized almost exclusively within the kidney tubulointerstitium where they are ideally placed to interact with proximal tubule epithelial cells (PTEC). We have previously shown that activated PTEC down-modulate autologous T lymphocyte and dendritic cell function. In this report, we extend these prior studies to describe PTEC-B cell interactions. METHODS: Stimulated B cells were cultured in the absence or presence of activated autologous human PTEC and monitored for proliferation, surface antigen expression, cytokine secretion and antibody (Ab) production. RESULTS: PTEC decreased B cell proliferative responses, whilst B cells cultured in the presence of PTEC displayed decreased levels of CD27, a marker of plasma B cells and memory cells. Interestingly, autologous PTEC also significantly decreased the number of B cells secreting both IgG and IgM and overall levels of Ab production. Transwell studies demonstrated that this modulation was primarily contact-dependent, and blocking studies with anti-PD-L1 led to partial restoration in Ab production. Further blocking studies targeting soluble HLA-G (sHLA-G) and IDO, two other immunoinhibitory molecules also up-regulated in our activated PTEC, demonstrated minor restoration of Ab responses. DISCUSSION: We report, for the first time, that PTEC are also able to modulate autologous B-cell phenotype and function via complex contact-dependent (PD-L1), soluble (sHLA-G) and intracellular (IDO) factors. We hypothesize that such mechanisms may have evolved to maintain peripheral immune-homeostasis, especially within the inflammatory milieu that exists within many kidney diseases.


Assuntos
Linfócitos B/fisiologia , Células Dendríticas/imunologia , Células Epiteliais/fisiologia , Túbulos Renais Proximais/fisiologia , Nefrite Intersticial/imunologia , Antígeno B7-H1/metabolismo , Comunicação Celular/fisiologia , Células Cultivadas , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Antígenos HLA-G/metabolismo , Humanos , Nefrite Intersticial/metabolismo , Linfócitos T/imunologia
8.
Kidney Int ; 87(6): 1153-63, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25587706

RESUMO

Chemokines play pivotal roles in tissue recruitment and retention of leukocytes, with CX3CR1 recently identified as a chemokine receptor that selectively targets mouse kidney dendritic cells (DCs). We have previously demonstrated increased tubulointerstitial recruitment of human transforming growth factor-ß (TGF-ß)-producing DCs in renal fibrosis and chronic kidney disease (CKD). However, little is known about the mechanism of human DC recruitment and retention within the renal interstitium. We identified CD1c+ DCs as the predominant source of profibrotic TGF-ß and highest expressors of the fractalkine receptor CX3CR1 within the renal DC compartment. Immunohistochemical analysis of diseased human kidney biopsies showed colocalization of CD1c+ DCs with fractalkine-positive proximal tubular epithelial cells (PTECs). Human primary PTEC activation with interferon-γ and tumor necrosis factor-α induced both secreted and surface fractalkine expression. In line with this, we found fractalkine-dependent chemotaxis of CD1c+ DCs to supernatant from activated PTECs. Finally, in comparison with unactivated PTECs, we showed significantly increased adhesion of CD1c+ DCs to activated PTECs via a fractalkine-dependent mechanism. Thus, TGF-ß-producing CD1c+ DCs are recruited and retained in the renal tubulointerstitium by PTEC-derived fractalkine. These cells are then positioned to play a role in the development of fibrosis and progression of chronic kidney disease.


Assuntos
Quimiocina CX3CL1/fisiologia , Células Dendríticas/fisiologia , Células Epiteliais/fisiologia , Túbulos Renais Proximais/citologia , Células Mieloides/fisiologia , Receptores de Quimiocinas/fisiologia , Adulto , Idoso , Antígenos CD1/análise , Receptor 1 de Quimiocina CX3C , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CX3CL1/análise , Quimiocina CX3CL1/metabolismo , Quimiotaxia , Células Dendríticas/química , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Fibrose/fisiopatologia , Glicoproteínas/análise , Humanos , Interferon gama/farmacologia , Túbulos Renais Proximais/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Células Mieloides/química , Receptores de Quimiocinas/análise , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
9.
PLoS One ; 9(1): e87345, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24475278

RESUMO

Interstitial fibrosis, a histological process common to many kidney diseases, is the precursor state to end stage kidney disease, a devastating and costly outcome for the patient and the health system. Fibrosis is historically associated with chronic kidney disease (CKD) but emerging evidence is now linking many forms of acute kidney disease (AKD) with the development of CKD. Indeed, we and others have observed at least some degree of fibrosis in up to 50% of clinically defined cases of AKD. Epithelial cells of the proximal tubule (PTEC) are central in the development of kidney interstitial fibrosis. We combine the novel techniques of laser capture microdissection and multiplex-tandem PCR to identify and quantitate "real time" gene transcription profiles of purified PTEC isolated from human kidney biopsies that describe signaling pathways associated with this pathological fibrotic process. Our results: (i) confirm previous in-vitro and animal model studies; kidney injury molecule-1 is up-regulated in patients with acute tubular injury, inflammation, neutrophil infiltration and a range of chronic disease diagnoses, (ii) provide data to inform treatment; complement component 3 expression correlates with inflammation and acute tubular injury, (iii) identify potential new biomarkers; proline 4-hydroxylase transcription is down-regulated and vimentin is up-regulated across kidney diseases, (iv) describe previously unrecognized feedback mechanisms within PTEC; Smad-3 is down-regulated in many kidney diseases suggesting a possible negative feedback loop for TGF-ß in the disease state, whilst tight junction protein-1 is up-regulated in many kidney diseases, suggesting feedback interactions with vimentin expression. These data demonstrate that the combined techniques of laser capture microdissection and multiplex-tandem PCR have the power to study molecular signaling within single cell populations derived from clinically sourced tissue.


Assuntos
Injúria Renal Aguda/fisiopatologia , Células Epiteliais/fisiologia , Regulação da Expressão Gênica/fisiologia , Túbulos Renais Proximais/citologia , Microdissecção e Captura a Laser/métodos , Reação em Cadeia da Polimerase Multiplex/métodos , Transdução de Sinais/fisiologia , Análise de Variância , Fibrose , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Túbulos Renais Proximais/fisiopatologia , Reação em Cadeia da Polimerase em Tempo Real
10.
Am J Physiol Renal Physiol ; 305(10): F1391-401, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24049150

RESUMO

Dendritic cells (DCs) play critical roles in immune-mediated kidney diseases. Little is known, however, about DC subsets in human chronic kidney disease, with previous studies restricted to a limited set of pathologies and to using immunohistochemical methods. In this study, we developed novel protocols for extracting renal DC subsets from diseased human kidneys and identified, enumerated, and phenotyped them by multicolor flow cytometry. We detected significantly greater numbers of total DCs as well as CD141(hi) and CD1c(+) myeloid DC (mDCs) subsets in diseased biopsies with interstitial fibrosis than diseased biopsies without fibrosis or healthy kidney tissue. In contrast, plasmacytoid DC numbers were significantly higher in the fibrotic group compared with healthy tissue only. Numbers of all DC subsets correlated with loss of kidney function, recorded as estimated glomerular filtration rate. CD141(hi) DCs expressed C-type lectin domain family 9 member A (CLEC9A), whereas the majority of CD1c(+) DCs lacked the expression of CD1a and DC-specific ICAM-3-grabbing nonintegrin (DC-SIGN), suggesting these mDC subsets may be circulating CD141(hi) and CD1c(+) blood DCs infiltrating kidney tissue. Our analysis revealed CLEC9A(+) and CD1c(+) cells were restricted to the tubulointerstitium. Notably, DC expression of the costimulatory and maturation molecule CD86 was significantly increased in both diseased cohorts compared with healthy tissue. Transforming growth factor-ß levels in dissociated tissue supernatants were significantly elevated in diseased biopsies with fibrosis compared with nonfibrotic biopsies, with mDCs identified as a major source of this profibrotic cytokine. Collectively, our data indicate that activated mDC subsets, likely recruited into the tubulointerstitium, are positioned to play a role in the development of fibrosis and, thus, progression to chronic kidney disease.


Assuntos
Antígenos CD1/análise , Antígenos de Superfície/análise , Quimiotaxia , Células Dendríticas/imunologia , Glicoproteínas/análise , Rim/imunologia , Lectinas Tipo C/análise , Células Mieloides/imunologia , Receptores Mitogênicos/análise , Insuficiência Renal Crônica/imunologia , Idoso , Biomarcadores/análise , Biópsia , Estudos de Casos e Controles , Contagem de Células , Citocinas/análise , Progressão da Doença , Feminino , Fibrose , Citometria de Fluxo , Humanos , Imunofenotipagem , Mediadores da Inflamação/análise , Rim/patologia , Masculino , Pessoa de Meia-Idade , Insuficiência Renal Crônica/patologia , Trombomodulina , Fator de Crescimento Transformador beta/análise
11.
Nephrol Dial Transplant ; 28(2): 303-12, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22610986

RESUMO

BACKGROUND: We have previously demonstrated that human kidney proximal tubule epithelial cells (PTEC) are able to modulate autologous T and B lymphocyte responses. It is well established that dendritic cells (DC) are responsible for the initiation and direction of adaptive immune responses and that these cells occur in the renal interstitium in close apposition to PTEC under inflammatory disease settings. However, there is no information regarding the interaction of PTEC with DC in an autologous human context. METHODS: Human monocytes were differentiated into monocyte-derived DC (MoDC) in the absence or presence of primary autologous activated PTEC and matured with polyinosinic:polycytidylic acid [poly(I:C)], while purified, pre-formed myeloid blood DC (CD1c(+) BDC) were cultured with autologous activated PTEC in the absence or presence of poly(I:C) stimulation. DC responses were monitored by surface antigen expression, cytokine secretion, antigen uptake capacity and allogeneic T-cell-stimulatory ability. RESULTS: The presence of autologous activated PTEC inhibited the differentiation of monocytes to MoDC. Furthermore, MoDC differentiated in the presence of PTEC displayed an immature surface phenotype, efficient phagocytic capacity and, upon poly(I:C) stimulation, secreted low levels of pro-inflammatory cytokine interleukin (IL)-12p70, high levels of anti-inflammatory cytokine IL-10 and induced weak Th1 responses. Similarly, pre-formed CD1c(+) BDC matured in the presence of PTEC exhibited an immature tolerogenic surface phenotype, strong endocytic and phagocytic ability and stimulated significantly attenuated T-cell proliferative responses. CONCLUSIONS: Our data suggest that activated PTEC regulate human autologous immunity via complex interactions with DC. The ability of PTEC to modulate autologous DC function has important implications for the dampening of pro-inflammatory immune responses within the tubulointerstitium in renal injuries. Further dissection of the mechanisms of PTEC modulation of autologous immune responses may offer targets for therapeutic intervention in renal medicine.


Assuntos
Comunicação Celular/fisiologia , Células Dendríticas/fisiologia , Células Epiteliais/fisiologia , Túbulos Renais Proximais/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Células Dendríticas/citologia , Células Epiteliais/citologia , Humanos , Imunidade/fisiologia , Túbulos Renais Proximais/citologia , Fagocitose/fisiologia , Linfócitos T/citologia , Linfócitos T/fisiologia
12.
Cancer Immunol Immunother ; 61(2): 169-179, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21874303

RESUMO

Immunotherapy is a promising new treatment for patients with advanced prostate and ovarian cancer, but its application is limited by the lack of suitable target antigens that are recognized by CD8+ cytotoxic T lymphocytes (CTL). Human kallikrein 4 (KLK4) is a member of the kallikrein family of serine proteases that is significantly overexpressed in malignant versus healthy prostate and ovarian tissue, making it an attractive target for immunotherapy. We identified a naturally processed, HLA-A*0201-restricted peptide epitope within the signal sequence region of KLK4 that induced CTL responses in vitro in most healthy donors and prostate cancer patients tested. These CTL lysed HLA-A*0201+ KLK4 + cell lines and KLK4 mRNA-transfected monocyte-derived dendritic cells. CTL specific for the HLA-A*0201-restricted KLK4 peptide were more readily expanded to a higher frequency in vitro compared to the known HLA-A*0201-restricted epitopes from prostate cancer antigens; prostate-specific antigen (PSA), prostate-specific membrane antigen (PSMA) and prostatic acid phosphatase (PAP). These data demonstrate that KLK4 is an immunogenic molecule capable of inducing CTL responses and identify it as an attractive target for prostate and ovarian cancer immunotherapy.


Assuntos
Antígenos de Neoplasias/metabolismo , Calicreínas/metabolismo , Fragmentos de Peptídeos/metabolismo , Neoplasias da Próstata/imunologia , Linfócitos T Citotóxicos/metabolismo , Adulto , Idoso , Antígenos de Neoplasias/imunologia , Proliferação de Células , Biologia Computacional , Células Dendríticas/imunologia , Feminino , Antígeno HLA-A2/metabolismo , Humanos , Epitopos Imunodominantes/genética , Calicreínas/imunologia , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/imunologia , Neoplasias da Próstata/patologia , Sinais Direcionadores de Proteínas/genética , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/patologia
13.
Nephrol Dial Transplant ; 26(5): 1483-92, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21045077

RESUMO

BACKGROUND: Renal proximal tubule epithelial cells (PTEC) respond and contribute to the pathological process in a range of kidney diseases. Within this disease setting, PTEC up-regulate surface antigens which may enable them to act as non-professional antigen-presenting cells and become targets for infiltrating T cells in the context of disease and allograft rejection. In order to define, for the first time, whether PTEC modulate immune responses within the autologous human system, we monitored their interaction with T and B cells in the presence of stimuli which mimic immunological signalling. METHODS: The expression of PTEC surface antigen in response to inflammatory mediators was monitored by flow cytometry. Purified T and B lymphocyte subsets and peripheral blood mononuclear cells were cultured in the presence or absence of autologous activated PTEC, and their responses to specific activators were monitored by proliferation, cytokine secretion and surface antigen expression. Some experiments were performed in the presence of blocking antibodies to PD-L1. RESULTS: The presence of activated primary autologous PTEC resulted in significantly decreased T- and B-cell proliferative responses, which were only partly mediated by programmed death ligand 1. This modulation was not induced by a decrease in activation markers or an increase in T regulatory cells but was accompanied by strong significant skewing of cytokine profiles. Significant decreases in gamma-interferon, interleukin-2 and tumour necrosis factor and increases in interleukin-4 were detected in the presence of PTEC, indicating that these cells induce a shift away from an inflammatory Th1 effector profile to a Th2 type profile. CONCLUSION: Human PTEC do modulate autologous immune responses. We hypothesize that such mechanisms may have developed to help dampen inflammatory responses and macrophage activation seen within kidney interstitium in many immune-mediated kidney diseases.


Assuntos
Linfócitos B/imunologia , Citocinas/imunologia , Interferon gama/farmacologia , Túbulos Renais Proximais/imunologia , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/farmacologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Proliferação de Células , Células Cultivadas , Citocinas/metabolismo , Citometria de Fluxo , Humanos , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
14.
Immunol Cell Biol ; 87(8): 606-14, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19546878

RESUMO

Dendritic cells differentiated from monocytes (MoDC) in the presence of GM-CSF and IL-15 (IL-15 MoDC) exhibit superior migration and cytotoxic T-lymphocyte (CTL) induction compared with MoDC differentiated in IL-4 and GM-CSF (IL-4 MoDC) and are promising candidates for DC immunotherapy. We explored the mechanisms by which IL-15 MoDC induce CTL. IL-15 MoDC expressed higher levels of CD40 and secreted high levels of TNF-alpha, but little or no IL-12p70 compared with IL-4 MoDC. Despite immuno-selecting monocytes to >97% purity before MoDC generation, a tiny population (0.2%) of natural killer (NK) cells was identified that was increased sevenfold during IL-15 MoDC, but not IL-4 MoDC differentiation. These NK cells produced high levels of IFN-gamma and were responsible for the enhanced CTL-inducing capacity of the IL-15 MoDC, but not for their increased expression of CD40 or secretion of TNF-alpha. Interestingly, a proportion of IL-15 MoDC were found to express the NK cell marker, CD56, but these did not secrete IFN-gamma. These data implicate a role for small percentages of NK cells in the enhanced capacity of IL-15 MoDC to induce tumour-specific CTL independent of IL-12p70.


Assuntos
Células Dendríticas/citologia , Células Dendríticas/imunologia , Interleucina-15/imunologia , Células Matadoras Naturais/imunologia , Monócitos/imunologia , Linfócitos T Citotóxicos/imunologia , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Diferenciação Celular , Células Cultivadas , Células Dendríticas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Interleucina-4/imunologia , Monócitos/citologia
15.
Prostate ; 66(2): 180-92, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16173035

RESUMO

BACKGROUND: Prostate cancer is one of the leading causes of cancer deaths in males and there are currently no effective treatments available for metastatic disease. Although recent clinical trials using dendritic cell (DC) based immunotherapy treatments have demonstrated safety, immunological responses, and some clinical efficacy, better vaccine delivery strategies need to be developed. We have undertaken the first detailed analysis of blood DC (BDC) subsets and their function in prostate cancer patients, with a view to utilizing immunoselected BDC for immunotherapy. METHODS: We enumerated the CD11c+CD1c+, CD11c+CD16+, and CD11c-CD123+ BDC subsets in whole blood of prostate cancer patients using a single platform TruCOUNT assay. These subsets were identified and purified using flow cytometry and immunomagnetic selection, and their functional capacity analyzed by costimulatory molecule expression, cytokine secretion, and antigen presenting ability. RESULTS: There were no significant differences in the number or composition of these subsets compared to healthy donors and these cells could be purified with equal efficiency from both groups. The prostate cancer patients BDC had similar levels of key costimulatory molecules and cytokine expression profiles, compared to healthy donors, and these were upregulated to the same extent, in response to exogenous stimuli. BDC from both groups were capable of eliciting allogeneic proliferative responses and inducing autologous CD4+ responses to naïve and recall antigens, and antigen-specific CD8+ responses to influenza matrix protein and prostate specific antigen. CONCLUSIONS: These results indicate that an immunoselected CD1c+ BDC preparation could provide a suitable vaccine delivery vehicle for future prostate cancer immunotherapy trials.


Assuntos
Células Dendríticas/imunologia , Neoplasias da Próstata/imunologia , Idoso , Apresentação de Antígeno , Antígenos CD/análise , Linfócitos T CD4-Positivos/imunologia , Estudos de Casos e Controles , Contagem de Células , Separação Celular , Citocinas/metabolismo , Citotoxicidade Imunológica , Células Dendríticas/citologia , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias da Próstata/sangue , Neoplasias da Próstata/terapia , Linfócitos T/fisiologia , Linfócitos T Citotóxicos/imunologia
16.
Blood ; 100(1): 184-93, 2002 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12070026

RESUMO

Platelet endothelial cell adhesion molecule-1 (PECAM-1/CD31) is an immunoglobulin-immunoreceptor tyrosine-based inhibitory motif (Ig-ITIM) superfamily member that recruits and activates protein-tyrosine phosphatases, SHP-1 and SHP-2, through its intrinsic ITIMs. PECAM-1-deficient (PECAM-1(-/-) ) mice exhibit a hyperresponsive B-cell phenotype, increased numbers of B-1 cells, reduced B-2 cells, and develop autoantibodies. In the periphery, there are reduced mature recirculating B-2 cells and increased B-1a cells within the peritoneal cavity. In addition, PECAM-1(-/-) B cells display hyperproliferative responses to lipopolysaccharide and anti-IgM stimulation and showed enhanced kinetics in their intracellular Ca(++) response following IgM cross-linking. PECAM-1(-/-) mice showed increased serum levels of IgM with elevated IgG isotypes and IgA antidinitrophenol antibody in response to the T-independent antigen, dinitrophenol-Ficoll. Finally, PECAM-1(-/-) mice developed antinuclear antibodies and lupuslike autoimmune disease with age.


Assuntos
Doenças Autoimunes/etiologia , Linfócitos B/imunologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/fisiologia , Receptores de Antígenos de Linfócitos B/efeitos dos fármacos , Animais , Formação de Anticorpos , Apoptose/efeitos dos fármacos , Autoanticorpos/biossíntese , Autoanticorpos/efeitos dos fármacos , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Medula Óssea/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Knockout , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/farmacologia , Receptores de Antígenos de Linfócitos B/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA