Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cold Spring Harb Protoc ; 2015(9): pdb.top070391, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26330629

ABSTRACT

Autophagy is a lysosomal catabolic pathway responsible for the degradation of cytoplasmic constituents. Autophagy is primarily a survival pathway for recycling cellular material in times of nutrient starvation, and in response to hypoxia, endoplasmic reticulum stress, and other stresses, regulated through the mammalian target of rapamycin pathway. The proteasomal pathway is responsible for degradation of proteins, whereas autophagy can degrade cytoplasmic material in bulk, including whole organelles such as mitochondria (mitophagy), bacteria (xenophagy), or lipids (lipophagy). Although signs of autophagy can be present during cell death, it remains controversial whether autophagy can execute cell death in vivo. Here, we will introduce protocols for detecting autophagy in mammalian primary cells by using western blots, immunofluorescence, immunohistochemistry, flow cytometry, and imaging flow cytometry.


Subject(s)
Autophagy , Cell Physiological Phenomena , Cytological Techniques/methods , Animals , Cells, Cultured , Humans , Mammals
2.
J Innate Immun ; 7(4): 375-91, 2015.
Article in English | MEDLINE | ID: mdl-25764971

ABSTRACT

Macrophages provide a bridge linking innate and adaptive immunity. An increased frequency of macrophages and other myeloid cells paired with excessive cytokine production is commonly seen in the aging immune system, known as 'inflamm-aging'. It is presently unclear how healthy macrophages are maintained throughout life and what connects inflammation with myeloid dysfunction during aging. Autophagy, an intracellular degradation mechanism, has known links with aging and lifespan extension. Here, we show for the first time that autophagy regulates the acquisition of major aging features in macrophages. In the absence of the essential autophagy gene Atg7, macrophage populations are increased and key functions such as phagocytosis and nitrite burst are reduced, while the inflammatory cytokine response is significantly increased - a phenotype also observed in aged macrophages. Furthermore, reduced autophagy decreases surface antigen expression and skews macrophage metabolism toward glycolysis. We show that macrophages from aged mice exhibit significantly reduced autophagic flux compared to young mice. These data demonstrate that autophagy plays a critical role in the maintenance of macrophage homeostasis and function, regulating inflammation and metabolism and thereby preventing immunosenescence. Thus, autophagy modulation may prevent excess inflammation and preserve macrophage function during aging, improving immune responses and reducing the morbidity and mortality associated with inflamm-aging.


Subject(s)
Aging/immunology , Autophagy/immunology , Macrophages/immunology , Microtubule-Associated Proteins/immunology , Aging/genetics , Aging/pathology , Animals , Autophagy/genetics , Autophagy-Related Protein 7 , Glycolysis/genetics , Glycolysis/immunology , Macrophages/pathology , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics
3.
J Clin Invest ; 124(9): 4004-16, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25083993

ABSTRACT

T cell senescence is thought to contribute to immune function decline, but the pathways that mediate senescence in these cells are not clear. Here, we evaluated T cell populations from healthy volunteers and determined that human CD8+ effector memory T cells that reexpress the naive T cell marker CD45RA have many characteristics of cellular senescence, including decreased proliferation, defective mitochondrial function, and elevated levels of both ROS and p38 MAPK. Despite their apparent senescent state, we determined that these cells secreted high levels of both TNF-α and IFN-γ and showed potent cytotoxic activity. We found that the senescent CD45RA-expressing population engaged anaerobic glycolysis to generate energy for effector functions. Furthermore, inhibition of p38 MAPK signaling in senescent CD8+ T cells increased their proliferation, telomerase activity, mitochondrial biogenesis, and fitness; however, the extra energy required for these processes did not arise from increased glucose uptake or oxidative phosphorylation. Instead, p38 MAPK blockade in these senescent cells induced an increase in autophagy through enhanced interactions between p38 interacting protein (p38IP) and autophagy protein 9 (ATG9) in an mTOR-independent manner. Together, our findings describe fundamental metabolic requirements of senescent primary human CD8+ T cells and demonstrate that p38 MAPK blockade reverses senescence via an mTOR-independent pathway.


Subject(s)
Autophagy/physiology , CD8-Positive T-Lymphocytes/physiology , MAP Kinase Signaling System/physiology , Multiprotein Complexes/physiology , TOR Serine-Threonine Kinases/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Adult , Autophagy-Related Proteins , Cellular Senescence , DNA Damage , Glycolysis , Humans , Mechanistic Target of Rapamycin Complex 1 , Membrane Potential, Mitochondrial , Membrane Proteins/physiology , Mitochondria/physiology , Oxidative Phosphorylation , Vesicular Transport Proteins/physiology
4.
Cell Mol Life Sci ; 70(1): 89-103, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22669258

ABSTRACT

Autophagy is a constitutive lysosomal catabolic pathway that degrades damaged organelles and protein aggregates. Stem cells are characterized by self-renewal, pluripotency, and quiescence; their long life span, limited capacity to dilute cellular waste and spent organelles due to quiescence, along with their requirement for remodeling in order to differentiate, all suggest that they require autophagy more than other cell types. Here, we review the current literature on the role of autophagy in embryonic and adult stem cells, including hematopoietic, mesenchymal, and neuronal stem cells, highlighting the diverse and contrasting roles autophagy plays in their biology. Furthermore, we review the few studies on stem cells, lysosomal activity, and autophagy. Novel techniques to detect autophagy in primary cells are required to study autophagy in different stem cell types. These will help to elucidate the importance of autophagy in stem cells during transplantation, a promising therapeutic approach for many diseases.


Subject(s)
Autophagy/physiology , Cell Differentiation , Cell Proliferation , Cellular Senescence , Models, Biological , Stem Cells/physiology , Animals , Humans , Lysosomes/physiology , Mice , Stem Cell Transplantation , Stem Cells/cytology
5.
Autophagy ; 8(4): 677-89, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22302009

ABSTRACT

Autophagy is a conserved constitutive cellular process, responsible for the degradation of dysfunctional proteins and organelles. Autophagy plays a role in many diseases such as neurodegeneration and cancer; however, to date, conventional autophagy detection techniques are not suitable for clinical samples. We have developed a high throughput, statistically robust technique that quantitates autophagy in primary human leukocytes using the Image stream, an imaging flow cytometer. We validate this method on cell lines and primary cells knocked down for essential autophagy genes. Also, using this method we show that T cells have higher autophagic activity than B cells. Furthermore our results indicate that healthy primary senescent CD8(+) T cells have decreased autophagic levels correlating with increased DNA damage, which may explain features of the senescent immune system and its declining function with age. This technique will allow us, for the first time, to measure autophagy levels in diseases with a known link to autophagy, while also determining the contribution of autophagy to the efficacy of drugs.


Subject(s)
Autophagy/immunology , Biological Assay/methods , Cellular Senescence/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Adult , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cells, Cultured , HEK293 Cells , Histones/metabolism , Humans , Mice , Middle Aged , Reproducibility of Results
6.
Autophagy ; 7(9): 1069-70, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21552009

ABSTRACT

The regulated lysosomal degradation pathway of autophagy prevents cellular damage and thus protects from malignant transformation. Autophagy is also required for the maturation of various hematopoietic lineages, namely the erythroid and lymphoid ones, yet its role in adult hematopoietic stem cells (HSCs) remained unexplored. While normal HSCs sustain life-long hematopoiesis, malignant transformation of HSCs or early progenitors leads to leukemia. Mechanisms protecting HSCs from cellular damage are therefore essential to prevent hematopoietic malignancies. By conditionally deleting the essential autophagy gene Atg7 in the hematopoietic system, we found that autophagy is required for the maintenance of true HSCs and therefore also of downstream hematopoietic progenitors. Loss of autophagy in HSCs leads to the expansion of a progenitor cell population in the bone marrow, giving rise to a severe, invasive myeloproliferation, which strongly resembles human acute myeloid leukemia (AML).


Subject(s)
Autophagy , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Hematopoietic System/pathology , Myeloid Cells/pathology , Animals , Autophagy-Related Protein 7 , Cell Proliferation , Hematopoietic System/metabolism , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/metabolism , Myeloid Cells/metabolism
7.
Cell Cycle ; 10(11): 1719-25, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21512311

ABSTRACT

Autophagy is a conserved cellular pathway responsible for the sequestration of spent organelles and protein aggregates from the cytoplasm and their delivery into lysosomes for degradation. Autophagy plays an important role in adaptation to starvation, in cell survival, immunity, development and cancer. Recent evidence in mice suggests that autophagic defects in hematopoietic stem cells (HSCs) may be implicated in leukemia. Indeed, mice lacking Atg7 in HSCs develop an atypical myeloproliferation resembling human myelodysplastic syndrome (MDS) progressing to acute myeloid leukemia (AML). Studies suggest that accumulation of damaged mitochondria and reactive oxygen species result in cell death of the majority of progenitor cells and, possibly, concomitant transformation of some surviving ones. Interestingly, bone marrow cells from MDS patients are characterized by mitochondrial abnormalities and increased cell death. A role for autophagy in the transformation to cancer has been proposed in other cancer types. This review focuses on autophagy in human MDS development and progression to AML within the context of the role of mitochondria, apoptosis and reactive oxygen species (ROS) in its pathogenesis.


Subject(s)
Autophagy/physiology , Leukemia, Myeloid, Acute/etiology , Myelodysplastic Syndromes/etiology , Bone Marrow Cells/pathology , Bone Marrow Cells/ultrastructure , Humans , Leukemia, Myeloid, Acute/pathology , Mitochondria , Myelodysplastic Syndromes/pathology , Reactive Oxygen Species
SELECTION OF CITATIONS
SEARCH DETAIL
...