Lamin in inflammation and aging

https://doi.org/10.1016/j.ceb.2016.03.004Get rights and content

Aging is characterized by a progressive loss of tissue function and an increased susceptibility to injury and disease. Many age-associated pathologies manifest an inflammatory component, and this has led to the speculation that aging is at least in part caused by some form of inflammation. However, whether or not inflammation is truly a cause of aging, or is a consequence of the aging process is unknown. Recent work using Drosophila has uncovered a mechanism where the progressive loss of lamin-B in the fat body upon aging triggers systemic inflammation. This inflammatory response perturbs the local immune response of the neighboring gut tissue and leads to hyperplasia. Here, we will discuss the literature connecting lamins to aging and inflammation.

Introduction

Chronic systemic inflammation without apparent infection in elderly humans is often referred to as the ‘inflammaging’ phenotype, and is primarily characterized by elevated levels of circulating pro-inflammatory cytokines [1, 2, 3, 4, 5, 6]. Epidemiological studies have found a strong correlation between the inflammaging phenotype and the presence of several aging-associated pathologies [7, 8, 9]. Genome-wide association studies have implicated several genes that function in immune, inflammatory and stress responses as being modifiers of longevity and health span [10].

Understanding the contribution of inflammation to aging has been pursued for many years with much effort aimed at identifying biomarkers of aging and cellular events that might serve as triggers for inflammatory responses. While conceptually straightforward, identifying cellular events has been very difficult, and is still largely correlative. Further, understanding the consequences of inflammation on aging presents its own challenges because, in part, many age-related diseases such as cardiovascular disease and osteoarthritis invoke strong inflammatory responses [11].

Recent studies have implicated the involvement of the nuclear intermediate filament proteins, the lamins, in aging-related inflammation. Here we will first introduce the general functions of lamins and then discuss the studies connecting these proteins to inflammation and aging.

Section snippets

Nuclear lamins

Nuclear lamins are classed as type V intermediate filaments. There are two lamin subtypes, A-type and B-type, which are distinguished by their protein sequences, physical properties and expression profiles. In humans, the A-type lamin is encoded by LMNA, while two separate genes, LMNB1 and LMNB2, encode the B-type lamins. In Drosophila, the subject of some discussion below, the A-type and B-type lamins (LAMC and LAM) are encoded by LamC and Lam, respectively.

Alternative splicing of the human

Lamin-A: aging and inflammation

Hutchinson-Gilford Progeria Syndrome (HGPS) is an exceptionally rare disorder that resembles premature aging. Most HGPS patients harbor the same LMNA mutation, a de novo C1824T change that enhances the use of a cryptic splice site and leads to the production of a permanently farnesylated form of lamin-A [31•, 32•]. This aberrant form of lamin-A, termed progerin, causes nuclear blebbing, down-regulation of some nuclear envelope proteins, accumulation of DNA damage and accelerated cellular

Lamin B: aging and inflammation

Human and mouse primary cell lines have a finite replicative lifespan (Hayflick limit) when cultured in vitro. This phenomenon, more commonly called replicative senescence, is characterized by cessation of cell division, increased secretion of inflammatory factors and changes in cell morphology and chromatin organization [45, 46]. Recent studies have found that the replicative senescence of cultured mammalian fibroblasts is accompanied by lamin-B1 reduction [47••, 48••, 49]. The induction of

Lamin-B may maintain tissue homeostasis by controlling gene expression

To gain insight into the role of lamin-B in maintaining the fat body organ, we performed RNA-seq to identify gene expression differences in fat bodies from young (5 days), old (50 days) and 5-day fat bodies that had LAM prematurely depleted by a tissue-specific RNAi [55••]. Gene ontology (GO) analyses revealed that age-associated LAM loss was characterized by changes in genes primarily belonging to immune, metabolic, proteolysis, and oxidative pathways (Figure 2a). Depletion of LAM in young fat

Conclusions and future considerations

Aging is multifactorial, and different theories that center on cellular damage have been proposed to explain this phenomenon [62]. While the relationship between the nuclear lamina and various forms of cellular damage has not been extensive explored, there are examples in the literature that suggest that protein homeostatic mechanisms, such as autophagy and the Ubiquitin Proteasome System, are particularly relevant. For instance, autophagy activation with either rapamycin or temsirolimus

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

This work was supported by the Ellison Medical Foundation and by NIH grants GM056312 and GM106023.

References (70)

  • C. Franceschi et al.

    Inflamm-aging. An evolutionary perspective on immunosenescence

    Ann N Y Acad Sci

    (2000)
  • A. Ahmad et al.

    Aging and inflammation: etiological culprits of cancer

    Curr Aging Sci

    (2009)
  • T. Fulop et al.

    Aging, immunity, and cancer

    Discov Med

    (2011)
  • M. Khatami

    Inflammation, aging, and cancer: tumoricidal versus tumorigenesis of immunity: a common denominator mapping chronic diseases

    Cell Biochem Biophys

    (2009)
  • F. Licastro et al.

    Innate immunity and inflammation in ageing: a key for understanding age-related diseases

    Immun Ageing

    (2005)
  • T.N. Akbaraly et al.

    Chronic inflammation as a determinant of future aging phenotypes

    CMAJ

    (2013)
  • T. Dechat et al.

    Nuclear lamins: major factors in the structural organization and function of the nucleus and chromatin

    Genes Dev

    (2008)
  • A.M. Pendas et al.

    Defective prelamin A processing and muscular and adipocyte alterations in Zmpste24 metalloproteinase-deficient mice

    Nat Genet

    (2002)
  • D.P. Corrigan et al.

    Prelamin A endoproteolytic processing in vitro by recombinant Zmpste24

    Biochem J

    (2005)
  • C.P. Maske et al.

    A carboxyl-terminal interaction of lamin B1 is dependent on the CAAX endoprotease Rce1 and carboxymethylation

    J Cell Biol

    (2003)
  • E.C. Schirmer et al.

    Nuclear membrane proteins with potential disease links found by subtractive proteomics

    Science

    (2003)
  • N. Korfali et al.

    The nuclear envelope proteome differs notably between tissues

    Nucleus

    (2012)
  • D.N. Simon et al.

    The nucleoskeleton as a genome-associated dynamic ‘network of networks’

    Nat Rev Mol Cell Biol

    (2011)
  • C. Smythe et al.

    Incorporation of the nuclear pore basket protein nup153 into nuclear pore structures is dependent upon lamina assembly: evidence from cell-free extracts of Xenopus eggs

    EMBO J

    (2000)
  • R. Foti et al.

    Nuclear architecture organized by Rif1 underpins the replication-timing program

    Mol Cell

    (2016)
  • Y. Kim et al.

    Mouse B-type lamins are required for proper organogenesis but not by embryonic stem cells

    Science

    (2011)
  • H. Chen et al.

    The nuclear lamina regulates germline stem cell niche organization via modulation of EGFR signaling

    Cell Stem Cell

    (2013)
  • C. Coffinier et al.

    Abnormal development of the cerebral cortex and cerebellum in the setting of lamin B2 deficiency

    Proc Natl Acad Sci U S A

    (2010)
  • C. Coffinier et al.

    Deficiencies in lamin B1 and lamin B2 cause neurodevelopmental defects and distinct nuclear shape abnormalities in neurons

    Mol Biol Cell

    (2011)
  • G. Bonne et al.

    Mutations in the gene encoding lamin A/C cause autosomal dominant Emery-Dreifuss muscular dystrophy

    Nat Genet

    (1999)
  • S. Shackleton et al.

    LMNA, encoding lamin A/C, is mutated in partial lipodystrophy

    Nat Genet

    (2000)
  • J.A. Damiano et al.

    Mutation of the nuclear lamin gene LMNB2 in progressive myoclonus epilepsy with early ataxia

    Hum Mol Genet

    (2015)
  • M. Eriksson et al.

    Recurrent de novo point mutations in lamin A cause Hutchinson-Gilford progeria syndrome

    Nature

    (2003)
  • A. De Sandre-Giovannoli et al.

    Lamin a truncation in Hutchinson-Gilford progeria

    Science

    (2003)
  • A. Chojnowski et al.

    Progerin reduces LAP2alpha-telomere association in Hutchinson-Gilford progeria

    Elife

    (2015)
  • Cited by (0)

    View full text