Next Article in Journal
Advances of Protein Palmitoylation in Tumor Cell Deaths
Previous Article in Journal
Regulation of Vascular Endothelial Growth Factor Signaling by Nicotine in a Manner Dependent on Acetylcholine-and/or β-Adrenergic-Receptors in Human Lung Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Multifaceted Roles of Lamins in Lung Cancer and DNA Damage Response

Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(23), 5501; https://doi.org/10.3390/cancers15235501
Submission received: 7 June 2023 / Revised: 15 November 2023 / Accepted: 17 November 2023 / Published: 21 November 2023
(This article belongs to the Special Issue New Insights to Cancer Therapy Resistance)

Abstract

:

Simple Summary

Lamins are a family of nuclear proteins consisting of A- and B-type lamins. As they are integral parts of the nuclear lamina, it was generally accepted that they have mainly structural functions. However, recent reports suggest a broader role for lamins in the context of cancer development and progression. Both the ratio of the different lamin subtypes as well as post-translational modifications can affect different cell states and processes including cellular stiffness, chromatin condensation, and regulation of the cell cycle. Here, we discuss the recent developments in lamin physiology and pathophysiology with a special focus on sometimes conflicting reports that describe the role of lamins in the context of lung cancer and the DNA damage response.

Abstract

Emerging evidence suggests that lamin functions are not limited to maintaining the structural integrity of the nucleus in eukaryotic cells but that these functions affect many facets of cancer biology. An increasing number of reports suggest that adaptive changes in the lamin subtype composition within the nuclear lamina could affect essential features of cancer development and aggressiveness. These include regulation of cellular stiffness and mobility as well as epithelial-to-mesenchymal transition (EMT), all of which directly impact the metastatic properties of cancer cells. Additionally, insights from studies on the physiological functions of lamins suggest that cancer cells could hijack the ability of lamins to modify chromatin accessibility, cell cycle regulation, and DNA damage response. Here, we present a comprehensive overview of the role of lamins in lung cancer and DNA damage response, which is commonly evoked by lung cancer therapies. Collectively, this information should help better understand the sometimes-conflicting reports on lamin functions in lung cancer as well as in other cancer types.

1. Introduction

Lamins are type V intermediate filament proteins that are best known for their scaffolding function in the nucleus of eukaryotic cells [1,2]. Lamins are encoded by the LMNA, LMNB1, and LMNB2 genes, giving rise to seven known lamin variants due to alternative splicing [3]. In physiological settings, lamins play important roles in maintaining the integrity of the nuclear envelope, regulating DNA replication and transcription, and organizing the chromatin structure [4,5]. Germline alterations in the lamin-encoding genes give rise to a multitude of disorders such as disturbed fat and skeletal homeostasis and syndromes that are summarized as laminopathies. These syndromes include cardiomyopathies, muscular dystrophies, and premature-aging-like syndromes such as the Hutchinson–Gilford progeria syndrome (HGPS). HGPS is caused by abnormal splicing of prelamin A, resulting in a shortened isoform that is referred to as lamin AΔ50, AΔ150, or progerin ([6], also reviewed in [7]). Lamin B-related diseases include lipodystrophy and brain disorders such as adult-onset autosomal dominant leukodystrophy (ADLD). While laminopathies are rare diseases, the underlying mutations provide insights into the function and organization of lamin proteins. Despite the fact that somatic mutations in the LMNA gene can occur in a variety of tumor types, these mutations are rare, and the mutation frequency does not exceed 3% in all cancer types analyzed by TCGA projects (Figure S1). In common cancers such as lung, prostate, and pancreatic tumors, lamin mutations occur in about 1% of cases, while the mutation rate was reportedly to be highest for LMNB2 in esophageal carcinomas (2.8%) [8]. However, given the overall low mutation rates of LMNA and LMNB genes, differences in mutation frequencies appear to be marginal (Figure S1). This, in turn, does not suggest a causal role for the mutation of a specific lamin-encoding gene in tissue-specific cancer development. By contrast, changes in the expression and localization of lamins have been frequently reported across cancer types (Table 1), indicating that lamins could contribute to the progression rather than to the onset of the disease. Changes in lamin expression and distribution will affect the integrity of the nuclear envelope, possibly contributing to chromosomal and genomic instability [9,10], which are hallmark features of cancer. Consequently, aberrant localization and expression, especially of lamin B proteins, have been associated with cancer development, aggressiveness, and metastasis (reviewed in [11]).
In this review, we first describe the physiological roles of lamins and put this into the context of the involvement of lamins governing genomic stability. We next analyze the differences of lamin expression reported in several types of cancer. Our main topic, lung cancer, is a model disease to further study lamins in the context of DNA damage response (DDR), chromatin organization, and their influence on EMT, as platinum-based chemotherapy and radiation, two mainstays in the therapy of lung cancer, both invoke DDR and DNA repair pathways. Although the interplay between these therapy-induced damage responses and changes in lamin expression or composition is not well understood, it might add to a better understanding of conflicting reports on lamins across cancer types.

2. Physiological Role of Lamins and Their Regulation during Cell Cycle and Cell Division

The nuclear lamina is a filamentous structure that determines the size, stability, and shape of the nucleus in eukaryotes [1]. The lamina consists mainly of type V intermediate filament proteins including A-type and B-type lamins [3]. A-type lamins are all encoded by the LMNA gene and can be divided into the four known alternative splice variants of lamin A, lamin C, lamin AΔex10, and lamin C2 [2,3,12]. B-type lamins are separately encoded by LMNB1 and LMNB2, which are translated into lamin B1 and lamin B2, respectively. A splice variant, lamin B3, is expressed from the LMNB2 gene using an alternative spermatid-specific promotor [13,14] (Figure 1B). A- and B-type lamins share the same domain structure consisting of an N-terminal head domain, a central α-helical rod domain including four coiled-coil segments, and a C-terminal IG-like tail domain (reviewed in [15]) (Figure 1A). Lamins form coiled-coil head-to-head dimers further connect to longitudinally linked head-to-tail polymers, which align laterally in an antiparallel manner to form tetrameric filaments [16]. Moreover, these tetrameric filaments are formed individually for A- and B-type lamins, but they build meshworks interacting with the nuclear envelope [17,18].
The phosphorylation of lamin domains prevents polymerization and thus impacts the lamin interactome (reviewed in [19]). While lamins contribute to nuclear integrity and stability, concerted lamin phosphorylation leading to depolymerization of the nuclear lamin structure and subsequent nuclear envelope breakdown is a prerequisite for cell division. Multiple phosphorylation sites within lamins are targeted by different kinases, including the cyclin-dependent kinase 1 (CDK1) [20], protein kinase A (PKA) [21], AKT (PKB) [22], protein kinase C (PKC) [21], mitogen-activated protein kinase MAPK [23] and casein kinase II [24] (Figure 1C).
During the interphase, when the nuclear lamina is attached to the nuclear envelope, lamin A/C interacts with emerin, an inner nuclear membrane protein [25], and both cooperate with nuclear myosin 1 (NM1) and actin to modulate chromatin organization. Loss of lamin A/C and emerin affects NM1 activity, which results in enhanced chromatin mobility, indicating that these interactions are important for maintaining chromosome positions during the interphase [26]. In prophase, DNA becomes condensed, and the nuclear envelope starts to break down (NEBD). During NEBD, the nuclear lamina becomes depolymerized and nuclear pore complexes (NPCs) dissociate, thus disrupting their interaction with chromatin. AKT and CDK1 were both reported to induce NEBD [20,27,28]. AKT was found to phosphorylate lamin A/C at Ser301 and Ser404 in C2C12 mouse myoblast cells [29,30] (Figure 1C). Bertacchini et al. [30] showed that the phosphorylation of prelamin A and lamin A/C by AKT marks both for degradation. Additionally, they reported AKT-mediated transcriptional control of LMNA, as prelamin A expression was downregulated in AKT-overexpressing cells. Complimentary results were obtained upon AKT knockdown and inhibition, which resulted in LMNA upregulation [31]. AKT activity peaks in the G2/M phase and drops right after mitosis [28], which recapitulates the coordinated and complex interactions underlying NEBD. Similarly, CDK1 is a prominent kinase phosphorylating lamins and NPC proteins to allow NEBD once CKD1 is activated by cyclin B1 [20,27]. Jeong et al. [32] showed that CDK1 activity is sufficient to disrupt the interaction of two parallel lamin dimers, which can be attenuated by casein kinase 1 (CK1) and glycogen synthase kinase-3 beta (GSK3β). The CDK1 complex targets several sites within different lamins, including lamin A/C (Thr19, Ser22, Ser392 and Ser404), lamin B1 (Ser23 and Ser393), and lamin B2 (Thr34, Ser37 and Ser405) (Figure 1C). All of these phosphorylation events contribute to lamin depolymerization and NEBD before the mitotic spindle assembles [20,33,34,35]. Upon NEBD, NPC components are distributed between the endoplasmic reticulum (ER) and cytoplasm [36]. Colocalization of lamin A/C and emerin is gradually lost in prometaphase, leading to the diffusion of lamins throughout the cell. By contrast, emerin remains detectable in the cytoplasm throughout mitosis, indicating a function of emerin for nuclear reassembly [37]. During cell division, B-lamins but not lamin A/C were reported to associate with the mitotic spindle. Using siRNA-induced depletion of lamin B1 or B2, Tsai et al. [38] showed increased spindle defects indicated by a significant increase in unfocused poles and/or a lack of chromosome alignment compared to control cells. These results indicate a functional role of B-type lamins during spindle assembly. After the alignment of chromosomes during metaphase, the NE starts to reassemble during ana- and telophase. With a gradual decrease of CDK1/CyclinB1 levels, the dephosphorylation of lamins by distinct phosphatases allows for re-polymerization and lamin A/C association with and around chromatin ([37], also reviewed in [39]). These observations indicate distinct roles of the different lamin types during cell division.

3. Contribution of Lamins to the Maintenance of Genomic Stability and Their Role in the DNA Damage Response

Aberrant DNA damage response (DDR) and evasion of senescence are hallmarks of cancer development and progression. DDR pathways include damage sensors, signal transducers, and effectors. Their deregulation contributes to genomic instability, which is associated with cancer onset and progression [40]. Cancer cells evade senescence either by activation of the enzyme telomerase (hTERT), which adds telomeric repeats to telomeres, or by utilizing the alternative lengthening of the telomer (ALT) pathway, which is based on homologous-directed repair (HDR) (reviewed in [41]). Tumor-specific strategies for stabilizing telomeres involve activation of hTERT by genomic rearrangements, mutational activation by point mutations, DNA amplification, and transcript fusions [42]. By contrast, the contribution of structural elements to telomere stability in cancer cells is not entirely understood. Crabbe et al. [43] reported the association of telomeres at the nuclear envelope during nuclear reassembly. Using an immuno-FISH approach, Pennarun et al. [44] demonstrated that DNA damage foci at telomeres were upregulated in lamin B1-overexpressing cells. Moreover, the integrity of the shelterin complex, which is essential for telomere protection, is affected by lamin B1 levels. Overexpression of lamin B1 induced mis-localization of the shelterin-complex members TRF2 and RAP1, subsequently leading to telomere instability [44]. This is functionally relevant, as a loss of TRF2 or RAP1 function increased the DDR and senescence [45,46,47]. However, the contribution of non-genetic stabilization of telomeres to telomere maintenance in cancer cells requires further investigation.
Another crucial factor that mediates proper DDR is the tumor suppressor p53 encoded by the TP53 gene (reviewed in [48]). Upon DNA damage, p53 becomes phosphorylated by the ataxia-telangiectasia mutated (ATM) kinase, and the checkpoint kinase 2 (Chk2) releases p53 from the mouse double minute 2 homolog (MDM2), which targets p53 for proteasomal degradation. Active p53 induces, among others, the expression of the cyclin-dependent kinase inhibitors p21 and p16, causing the cell cycle arrest or cellular senescence, depending on the context (reviewed in [49]). Interestingly, Yoon et al. [50] reported a direct interaction between p53 and lamin A/C, but not lamin B, using in vitro binding assays. Moreover, increased p53 levels preferentially stabilize lamin A/C, which promotes nuclear deformation and induction of p16 [50]. In the same manuscript, the authors speculated that lamin A/C stabilization might be sufficient to induce senescence [50]. By contrast, Chuang et al. [51] reported that inactivation of the focal adhesion kinase (FAK) leads to upregulation of p53, which induced cellular senescence while downregulating lamin A/C. Additionally, they suggested FAK-mediated regulation of lamin A/C expression as a means to stabilize the nuclear organization and to activate anti-senescence programs [51]. However, Golubovskaya et al. [52] have shown that this interaction may be bi-directional, as p53 binds to the FAK promotor, thus inhibiting its activity. By analyzing the effect of lamin A/C depletion on the nuclear envelope and focal adhesions, Goelzer et al. [53] found upregulated FAK and AKT levels in LMNA-depleted cells and suggested this upregulation as compensation for cell softening upon lamin A/C deficiency. Together, these results indicate FAK as another important regulator in p53-mediated senescence through its interaction with lamin A/C and p53. At present, it remains unclear how these mutual interactions between FAK, p53, and lamin A/C are coordinated to regulate DDR and cellular senescence.
In addition, lamin B1 was reported as a p53 target. Loss of p53 leads to upregulation of lamin B1 and other members of the nuclear lamina, including the nuclear pore complex 210 (Nup210). Both coding genes share genomic binding sites with p53, which was also functionally relevant in a pancreatic ductal adenocarcinoma cell line [54]. Consistent with these findings, Lämmerhirt et al. [55] reported that loss of lamin B1 induces senescence in melanoma cells, while Freund et al. [56] found that lamin B1 down-regulated upon the activation of p53 or pRB/p16INK4a. Following this observation, Onorati et al. [57] recently showed that different means to induce senescence, including replication exhaustion, expression of HRAS-V12, etoposide-mediated DNA damage, or ionizing irradiation, all resulted in decreased lamin B1 levels. These results indicate that p53, with or without the involvement of FAK, has opposing effects on the stabilization of lamin A/C and lamin B1, which thus may be an important switch regulating cellular senescence upon DNA damage.
The persistence of DNA double-strand breaks (DSBs), which are lethal when unrepaired, poses a dangerous threat to genomic integrity. Here, p53-binding protein 1, 53BP1 [58], is an important factor facilitating DSB repair, among others by protecting DNA ends from resection [59]. Structurally, the tudor and the ubiquitination-dependent recruitment (UDR) domains of 53BP1 enable its interaction with DSB-specific histone marks, H4K20me2, and ubiquitinated lysine 15 on H2A-type histones (H2A-K15ub) [60,61], thus establishing 53BP1 as a reader of DNA damage. Gibbs-Seymour et al. [62] described that lamin A/C specifically interacts with 53BP1s via the tudor domain. Here, irradiation-induced DNA DSBs resulted in a decreased interaction of lamin A/C and 53BP1 [62]. This interaction is functionally relevant, as lamin A/C depletion also resulted in a loss of the 53BP1 protein and concomitantly accumulated DNA damage [63]. Interestingly, proximity ligation assays also suggested a direct interaction between 53BP1 and lamin B1, which reportedly also stabilized the 53BP1–lamin A/C complex [64]. However, overexpression of lamin B1 resulted in impaired 53BP1 recruitment to DNA damage sites, decreased NHEJ, and thus resulted in the accumulation of unrepaired DNA DSB. Taken together, these results indicate distinct roles for lamin A/C and lamin B1 during DNA damage response and DNA double-strand repair that need further investigation to specify their function.
Of note, mutant LMNAD300N predisposing to dilated cardiomyopathy in humans was shown to induce an aberrant DNA damage response involving ATM and p53, which could be partly rescued by p53 deletion in transgenic mice [65]. It is thus tempting to speculate that interfering with p53 and the DDR in laminopathies might ameliorate the pathophysiological consequences of lamin mutations. Additionally, Gonzalez-Suarez et al. [63] showed that interaction of lamin A/C with the DDR machinery is not restricted to its interaction with p53 pathway members, but can also involve telomeres. In LMNA−/− mouse embryo fibroblasts (MEFs), telomeres were significantly shorter than in LMNA-wt MEFs, which correlated with a decrease in H4K20me3, a histone mark of telomeric heterochromatin. Additionally, loss of A-type lamins caused nuclear decompartmentalization of telomeres and hindered the ability to process dysfunctional telomeres [63]. These reports indicate that lamin A/C and B1 target distinct aspects of telomere stability, DDR, and senescence that determine their impact on pathophysiology.

4. Role of Lamins in Lung Cancer

Lung cancer is divided into two subtypes, namely the more frequently occurring non-small-cell lung cancers (NSCLC) and small-cell lung cancers (SCLC), which account for roughly 85% and 15% of all lung cancers, respectively. Depending on the subtype, varying levels of lamin A/C were detected in lung tumor cells. SCLC expresses LMNA at lower levels compared to NSCLC tumor tissues [66,67]. In pre-clinical models, conflicting results have been reported by Stefanello et al. [68], who found decreased lamin A/C levels in the lung adenocarcinoma (LUAD) cell line A549, while Rubporn et al. [69] reported LMNA overexpression in the same cell line. Furthermore, Hu et al. [70] reported that lamin A/C contributes to acquired resistance to erlotinib in EGFR-mutant NSCLC. They analyzed cytoskeletal changes and found increased nuclear deformation upon the modulation of LMNA expression. Based on these findings, they suggested that reduced LMNA expression contributes to higher nuclear plasticity. As for lamin A/C, different effects of lamin B1 on lung cancer biology were reported: Jia et al. [67] identified decreased lamin B1 expression as a lung cancer-promoting factor, whereas two reports by Li et al. [71] and Li et al. [72] suggested that lamin B1 overexpression correlated with advanced-stage NSCLC. Interestingly, lamin B2 expression levels were unanimously correlated with a higher clinical stage [73,74,75]; however, the mechanism underlying lamin B2 regulation still needs clarification. Taken together, dysregulation of the different lamin proteins is observed in lung cancer, but the impact derived thereof remains controversial. However, three eminent functions of lamins have been identified that seem to depend on the expression of the different lamin subtypes and that can be hijacked in lung cancer cells: 1. cell cycle regulation induced by receptor tyrosine kinase or protein kinase signaling that affects lamin polymerization by phosphorylation; 2. chromatin organization and compactness that can impact transcriptional activity; and 3. cellular stiffness and mobility that can be molecularly linked to lamin interaction with proteins regulating EMT. Beyond these central processes, lamins might also contribute to DNA damage response and repair pathways ([76], also reviewed in [77]).

4.1. Role of Lamin Dysregulation in DNA Damage Response, Cell Cycle Transition, and DNA Repair

The facts that nuclear integrity mediated by lamins controls cell fate and that rupture of the nuclear lamina is a catastrophic event causing programmed cell death are well acknowledged. However, in genomically unstable cancer cells, controlling functions of lamins can be overruled and hijacked. A prime example is the formation of lamin B1-containing micronuclei in lung cancer cells that form around missegregated chromosomes during mitotic exit, which in turn can contribute to genomic instability and even potentiate it [76]. Still, this structural function of lamins is only one side of the coin, as lamins can also interact with proteins that directly control cell cycle and proliferation.
Rubporn et al. [69] found that lamin A/C and several co-regulated proteins including KAP1 (TIF1β, TRIM28) were overexpressed in the A549 lung cancer cell line compared to MRC-5 normal lung fibroblast cells. Interestingly, Neumann-Staubitz et al. [33] identified KAP1 as an interactor of the Ig-domain of lamin C using genetically encoded crosslinkers. KAP1 is involved in cell survival after DNA damage and its phosphorylation by ATM, ATR, and DNA-PK recruits KAP1 to DNA strand breaks [78]. Furthermore, KAP1 is part of the transcription intermediary factor 1 (TIF1) sub-group, whose members possess a bromodomain (Bromo) and a plant homeodomain (PHD), which are common reader domains for the epigenetic code of histone post-translational modifications [79,80]. KAP1 was found to directly interact with MDM2, leading to the recruitment of the p53-HDAC1 complex that results in p53 inhibition and impaired apoptosis [81,82]. Consequently, the interaction of KAP1 with lamin C and its upregulation indicates a function of lamin A/C in cell survival in A549 cells, which needs to be verified in additional model systems.
In addition to lamin A/C, B-type lamins were reported to be deregulated in lung cancer. Immunohistology and biochemical analyses suggested that lamin B2 is upregulated in NSCLC and promotes aggressiveness by inducing or interacting with KLF16, MCM7, and G9a [73,74,75]. Here, expression of G9a depended on both lamin B2 and cyclin D1 expression, contributing to enhanced cell migration and cell cycle progression. Ma et al. [74] reported cell proliferation after overexpression of LMNB2 and impaired G1/S cell cycle progression upon lamin B2 knockdown with concomitantly decreased levels of cyclin D1 and cyclin E1. Interestingly, cyclin B1 expression and G2/M phase transition seemed to be unaffected by lamin B2 levels, suggesting that lamin B2 specifically targets G1/S transition. However, overexpression of another lamin B2 interaction partner, KLF16, in H1299 and H1975 LUAD cell lines resulted in lamin B2 upregulation accompanied by enhanced proliferation and migration. Here, the fraction of cells in the G0/G1 phase decreased while G2/M cells were increased. Taken together, these results indicate that lamin B2 expression levels can impact cell cycle progression via different mechanisms.

4.2. Lamin Dysregulation and Chromatin Organization in Lung Cancer

The interaction between chromatin and the nuclear lamina determines, at least in part, chromatin structure and accessibility [83]. At present, it is not known how the specificity of these interactions is regulated and how this physiological role of creating permissive or restrictive environments for gene transcription is hijacked in cancer cells. As described before, inactivation of FAK was reported to induce cellular senescence and p53 upregulation [51]. Recently, Chuang et al. [84] further investigated the mechanism of FAK-diminished senescence involving the enhancer of zeste homolog 2 (EZH2), which is a histone methyltransferase within the PRC2-repressor complex regulating H3K27 methylation and gene silencing [85,86]. Inhibition of EZH2 was previously reported to be involved in the onset of senescence in different cancer types such as triple-negative breast cancer or pancreatic cancer [87,88]. Furthermore, it was shown that FAK inhibition leads to senescence in lung cancer involving the downregulation of EZH2, suggesting a functional correlation of EZH2 and FAK in non-small cell lung cancer [84]. Interestingly, lamin B1 was reported to specifically interfere with the function of EZH2 [67]. By depleting lamin B1, Jia et al. [67] showed that chromatin-bound EZH2 was reduced concomitant with a decrease in H3K27me3. They suggested that lamin B1 is important for H3K27me3-mediated chromatin accessibility and transcriptional regulation [67]. These results and the aforementioned in vitro reports underscore that lamin A/C and lamin B1 could both contribute to the dysregulation of cellular senescence and epigenetic modifiers involved in cell division in lung cancer.

4.3. Impact of Lamin Dysregulation on Cellular Mobility, Plasticity, and EMT in Lung Cancer Cells and Lung Metastases

The plasticity and dynamic regulation of lamin proteins in lung cancer cells are highly debated and poorly understood. Hu et al. [70] investigated the consequences of downregulating LMNA in lung cancer cells on EMT by quantifying the ratio of E-cadherin and vimentin. In cells with reduced LMNA levels, E-cadherin was downregulated and vimentin was upregulated, indicating EMT accompanied by enhanced migration and invasion [70]. In a corresponding approach, Jia et al. [67] showed that depletion of lamin B1 in normal mouse lung epithelial cells induced the downregulation of E-cadherin and upregulation of the mesenchymal markers fibronectin, vimentin, and N-cadherin. Moreover, loss of lamin B1 led to upregulation of the proto-oncogene RET and its coreceptor GFRA1 together with upregulation of the MAP kinase p38. RET is a member of the receptor tyrosine kinase family and its activation stimulates several pathways, including the MAPK and PI3K/AKT signaling pathways (reviewed in [89]), which are involved in EMT (reviewed in [90]). Therefore, loss of lamin B1 could contribute to EMT via the activation of RET and its downstream function in the MAPK and PI3K/AKT pathways. On the contrary, Li et al. [71] reported that lamin B1 overexpression in LUAD was sufficient to activate the AKT/pAKT pathway. Depletion of lamin B1 slowed the tumor growth of A549 cells concomitant with decreased phosphorylation of AKT. They suggested that lamin B1 regulates cell proliferation in an AKT-dependent manner and that lamin B1 may function as an oncogene in LUAD. Interestingly, Pascual-Reguant et al. [91] showed that lamin B1 expression is essential for the acquisition of mesenchymal traits during EMT in the epithelial-like mouse cell line NMuMG. However, the role of lamin B1 in the context of EMT needs to be consolidated, as lamin B1 orchestrates signals from different pathways including AKT signaling, which could induce cell type-specific responses.

5. Discussion

This review summarizes the functions of lamins in eukaryotic cells and focuses on the role of the lamin protein family in DNA damage response in lung cancer. We integrate current knowledge on lamin functions beyond acting as structural components of the cell nucleus (summarized in Figure 2). However, there is currently no clear picture emerging especially for the role of lamin B1 in lung cancer, which was reported to have either oncogenic or tumor-suppressive functions [67,71,72]. As lamin deregulation is commonly noted across cancer entities with sometimes conflicting observations, we also provide an overview of the currently available distribution of lamin A/C, B1, and B2 expression in diverse types of cancers, both in cell lines and patient-derived tissue (Table 1, Table 2 and Table 3). Notably, changes in lamin composition can also be found in many other tumor entities including the most frequent cancers of the breast [9,92], skin [55,93], and also digestive tract cancers [94,95,96,97,98]. On a general note, lamin expression is highly variable between cancers and different studies on lamins in large patient cohorts reported discrepancies concerning the clinical course and lamin expression (Table 1, Table 2 and Table 3). For instance, lamin A/C upregulation was reported in many tumor types such as prostate cancer, colorectal cancer, and glioblastoma, while lamin A/C was underrepresented in small-cell lung cancer, breast cancer, ovarian cancer, Ewing sarcoma, gastric cancer, and colon cancer (Table 1 and references therein). Patients with colorectal cancer (n = 656) expressing lamin A/C were reported to have a higher risk of disease progression than those without lamin A/C expression [95]. By contrast, loss of lamin A/C expression was associated with disease recurrence in another study involving colon cancer patients (n = 370). This discrepancy is also reflected in several studies, which reported different results when analyzing lamin A/C expression in NSCLC. Here, lamin A/C expression was altered in vitro in tumor cells compared to non-cancer cells and normal lung fibroblasts [68,69], whereas Jia et al. [67] found no differences in lamin A/C expression in a series of primary tumors by using immunohistochemistry. Moreover, different reports showed that either lamin B1 loss or lamin B1 overexpression promotes NSCLC aggressiveness [67,71,72]. Opposingly, lamin B2 overexpression unambiguously surfaced as an unfavorable event in lung cancer [73,74,75]. To sum this part up, the data availability concerning the biological function and the prognostic value of lamin expression is unsatisfactory except for lamin B2, which might serve as an indicator of aggressive disease courses.
To analyze if the conflicting results regarding lamin expression are due to different technical approaches, we inspected the methodology used in the respective studies (Tables S1–S3). These differences might arise when RNA levels and protein levels are compared or when technologies and platforms have limited overlap. However, methodological differences between studies could not fully explain the conflicting results. Additionally, the choice of reference groups (e.g., normal tissues, less-aggressive tumor types) did not correlate with the reported regulation of the lamins. However, as lamin isoform expression could be variable and differ between proliferating and resting cells, the ratios of lamin expression and correction for cell proliferation (e.g., by staining for Ki-67) would allow for a more precise view on the role of the differential expression of lamins across tumor types. Another important caveat has also recently been postulated by Roncato and colleagues [99], who showed that lamin A/C deficiency resulted in deformable nuclei, which enhanced migratory capacity but not metastasis in vivo. These results demonstrate that the readouts for evaluating functional consequences must be standardized when examining the complex interplay between nuclear membrane rigidity, migratory capacity, invasiveness, and metastatic ability. This also holds when lamins are analyzed in the context of resistance-associated EMT [70]. While lamins have a significant impact on the deformability of the cell nucleus and thus could favor the migration of potentially metastasizing cells, it is an oversimplification to assume that reduced A-type lamins automatically increase the metastatic ability of circulating cancer cells. Moreover, Nishikawa et al. [100] proposed an impact of distinct LMNA transcript variants on cancer cell proliferation. They focused on the LMNA transcript variant 6 (LMNA-V6), whose promotor region forms G-quadruplexes, thus increasing its transcriptional activity. Upregulation of LMNA-V6 led to the downregulation of other lamin A transcripts. In addition, LMNA-V6 expression also correlated with impaired p53 levels, suggesting a tumor-promoting function of this isoform [100]. Again, lamin B2 is the only lamin subtype whose expression is consistently annotated as pro-tumorigenic and correlates with poor patient prognosis across entities (Table 3 and Table S3). As for other cancer types, reports on lung cancer suggest that lamins are effectors of oncogenic processes that pave the way for malignant progression (Figure 2). However, a causal role for cancer development initiated by altered lamins was not established. This notion is also corroborated by the low frequency of lamin mutations in lung cancer and other diverse tumor types (Figure S1). Therefore, it remains to be determined if expression shifts or rather a specific combination of different lamin subtypes or transcript variants contribute to pro- or anti-tumorigenic properties of the lamin network. Data analyses of lamin subtypes should also consider transcript variants so as to dissect their impact on cancer development and progression. Moreover, the interaction between different lamins and their transcript variants that are governed by phosphorylation-dependent polymerization should be further investigated. Clearly, these ambiguous results call for a unified description of the methodology used in the manuscripts that deal with the role of lamins in cancer. Harmonization of protocols will then contribute to a deeper understanding of the role of lamins in cancer biology and progression.
Table 1. Lamin A/C expression levels in diverse types of cancer.
Table 1. Lamin A/C expression levels in diverse types of cancer.
CancerCell LineLamin ExpressionReference
RegulationReference ParameterImpact
Ovarian Cancern = 1↑*Ovarian epithelial cells/[101]
Non-small cell lung
cancer
n = 1
n = 1

Non-cancer cells
Normal lung fibroblast cells
/
/
[68]
[69]
Small cell lung cancern = 3NSCLC cell lines/[66]
Osteosarcoman = 2Low aggressive cell lineMigration[102]
CancerTumor
Tissue
Lamin ExpressionReference
RegulationReference ParameterImpact
Prostate cancern = 46
n = 94
n = 94
↑*
↑*
Benign sample
High- vs. low-grade tissue
/
/
Advanced stage
Invasion
[103]
[103]
[104]
Glioblastoma multiformen = 152/Poor prognosis[105]
Colorectal cancern = 656/Poor prognosis[97]
Small cell lung cancern = 20
n = 33

NSCLC
NSCLC
/
/
[67]
[66]
Breast cancern = 115Non-cancerous tissuePoor prognosis[9]
Ovarian cancern = 108Ovarian surface epithelial cells/[10]
Ewing Sarcoman = 64Metastatic vs. primary tumorPoor prognosis[106]
Gastric cancern = 52Normal tissuePoor prognosis[98]
Colon cancern = 35
n = 370

Colonic mucosa
No recurrence
/
Recurrence
[96]
[95]
Overview of lamin A/C expression in tumor cell lines and tumor tissue as indicated. “Reference parameter” refers to the tissue or cell that served as a reference for lamin A/C expression. “n”: number of analyzed cells lines/tumor tissues; “↑”: increased expression; “↑*”: increased expression only for lamin A/C; “↓”: decreased expression.
Table 2. Lamin B1 expression levels in diverse types of cancer.
Table 2. Lamin B1 expression levels in diverse types of cancer.
CancerCell LineLamin ExpressionReference
RegulationReference ParameterImpact
Ovarian cancern = 11Ovarian surface epithelial cells/[10]
Hepatocellular carcinoman = 5MIHA and HepG2/[107]
Melanoman = 6
n = 1
n = 2

↑*
↓*
Normal epidermal melanocytes
/
/
/
Migration
Senescence
[55]
[93]
[55]
Prostate cancern = 1↑*/Proliferation & invasion[108]
Pancreatic cancern = 2↓*/Migration & Invasion[109]
CancerTumor
Tissue
Lamin ExpressionReference
RegulationReference ParameterImpact
Ovarian cancern = 27Benign tumors/[110]
Pancreatic cancern = 5Normal pancreatic tissuePoor prognosis[109]
Hepatocellular carcinoman = 39
n = 364 + 229

Cirrhosis and non-diseased tissue
Low metastasis score group
/
Poor prognosis
[107]
[111]
Clear cell renal cell carcinoman = 622Normal renal tissuePoor prognosis[112]
Non-small cell lung
cancer
n = 483
n = 139

Normal lung tissue
Normal lung tissue
Poor prognosis
Advanced stage
[71,72]
[67]
Small cell lung cancern = 22Normal lung tissue/[67]
Breast cancern = 115Non-cancerous tissuePoor prognosis[9]
Colon cancern = 35Normal colonic mucosa/[96]
Overview of lamin B1 expression in tumor cell lines and tumor tissue as indicated. “Reference parameter” refers to the tissue or cell that served as a reference for lamin B1 expression. “n”: number of analyzed cells lines/tumor tissues; “↑”: increased expression; “↑*”: increased expression only for lamin A; “↓”: decreased expression. “↓*”: decreased expression only for lamin A.
Table 3. Lamin B2 expression levels in diverse types of cancers.
Table 3. Lamin B2 expression levels in diverse types of cancers.
CancerCell LineLamin ExpressionReference
RegulationReference ParameterImpact
Colorectal cancern = 5Normal colon epithelial cell lineProliferation[94]
CancerTumor
Tissue
Lamin ExpressionReference
RegulationReference ParameterImpact
Non-small cell lung
cancer
n = 526
n = 20
n = 150
n = 135



Normal lung sample
Adjacent normal lung tissue
Low-grade tumor
Normal lung tissue
Poor prognosis
/
Poor prognosis
Poor prognosis
[73]
[74]
[74]
[75]
Breast cancern = 82
n = 1085

Normal tissue
Corresponding normal tissue
Poor prognosis
/
[92]
[92]
Ovarian cancern = 27Benign tumor/[110]
Colorectal cancern = 226Adjacent tissuePoor prognosis[94]
Overview of lamin B2 expression in tumor cell lines and tumor tissue as indicated. “Reference parameter” refers to the tissue or cell that served as a reference for lamin B2 expression. “n”: number of analyzed cells lines/tumor tissues; “↑”: increased expression; “↓”: decreased expression.

6. Conclusions

While lamins are indispensable for the structural integrity of the nucleus in normal cells, aberrant lamin subtype expression and composition as well as the lamin interactome in lung cancer and other tumor types is not yet fully understood. Even the role of the same lamins in and between cancer entities remains debatable. Nevertheless, it is evident that lamins orchestrate a variety of cellular cancer-related processes beyond their structural functions. In lung cancer, lamin composition and subtype expression govern the DNA damage response that is invoked by current therapies. The impact of lamins on therapy response and cancer progression deserves further investigation.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers15235501/s1, Figure S1: Mutation frequencies for LMNA, LMNB1 and LMNB2 genes across common cancer entities. Data were obtained from the “The Cancer Genome Atlas” (TCGA) projects available at the Genomic Data Commons Portal (https://portal.gdc.cancer.gov, accessed on 6 November 2023); Table S1: Methodological details for Table 1: Lamin A/C expression levels in diverse types of cancer, Table S2: Methodological details for Table 2: Lamin B1 expression levels in different types of cancer, Table S3: Methodological details for Table 3: Lamin B2 expression levels in different types of cancers.

Author Contributions

Conceptualization, J.J., S.O. and A.S.; writing—original draft preparation, J.J.; writing—review and editing, S.O. and A.S.; visualization, J.J. and S.O.; supervision, A.S.; project administration, A.S.; funding acquisition, A.S. and S.O. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by a grant from the DFG (Deutsche Forschungsgemeinschaft) to A.S. (GRK 2762, subproject L3) and from the Daimler and Benz Foundation to S.O. (32-05/19).

Acknowledgments

Figures were partially created with BioRender.com (accessed on 7 November 2023).

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Lammerding, J.; Fong, L.G.; Ji, J.Y.; Reue, K.; Stewart, C.L.; Young, S.G.; Lee, R.T. Lamins A and C but not lamin B1 regulate nuclear mechanics. J. Biol. Chem. 2006, 281, 25768–25780. [Google Scholar] [CrossRef]
  2. Fisher, D.Z.; Chaudhary, N.; Blobel, G. cDNA sequencing of nuclear lamins A and C reveals primary and secondary structural homology to intermediate filament proteins. Proc. Natl. Acad. Sci. USA 1986, 83, 6450–6454. [Google Scholar] [CrossRef]
  3. Machiels, B.M.; Zorenc, A.H.; Endert, J.M.; Kuijpers, H.J.; van Eys, G.J.; Ramaekers, F.C.; Broers, J.L. An alternative splicing product of the lamin A/C gene lacks exon 10. J. Biol. Chem. 1996, 271, 9249–9253. [Google Scholar] [CrossRef]
  4. Bouvier, D.; Hubert, J.; Seve, A.P.; Bouteille, M. Characterization of lamina-bound chromatin in the nuclear shell isolated from HeLa cells. Exp. Cell Res. 1985, 156, 500–512. [Google Scholar] [CrossRef]
  5. Sullivan, T.; Escalante-Alcalde, D.; Bhatt, H.; Anver, M.; Bhat, N.; Nagashima, K.; Stewart, C.L.; Burke, B. Loss of a-Type Lamin Expression Compromises Nuclear Envelope Integrity Leading to Muscular Dystrophy. J. Cell Biol. 1999, 147, 913–920. [Google Scholar] [CrossRef]
  6. Eriksson, M.; Brown, W.T.; Gordon, L.B.; Glynn, M.W.; Singer, J.; Scott, L.; Erdos, M.R.; Robbins, C.M.; Moses, T.Y.; Berglund, P.; et al. Recurrent de novo point mutations in lamin A cause Hutchinson-Gilford progeria syndrome. Nature 2003, 423, 293–298. [Google Scholar] [CrossRef]
  7. Wong, X.; Stewart, C.L. The Laminopathies and the Insights They Provide into the Structural and Functional Organization of the Nucleus. Annu. Rev. Genomics Hum. Genet. 2020, 21, 263–288. [Google Scholar] [CrossRef]
  8. Tate, J.G.; Bamford, S.; Jubb, H.C.; Sondka, Z.; Beare, D.M.; Bindal, N.; Boutselakis, H.; Cole, C.G.; Creatore, C.; Dawson, E.; et al. COSMIC: The Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res. 2019, 47, D941–D947. [Google Scholar] [CrossRef]
  9. Wazir, U.; Ahmed, M.H.; Bridger, J.M.; Harvey, A.; Jiang, W.G.; Sharma, A.K.; Mokbel, K. The clinicopathological significance of lamin A/C, lamin B1 and lamin B receptor mRNA expression in human breast cancer. Cell. Mol. Biol. Lett. 2013, 18, 595–611. [Google Scholar] [CrossRef]
  10. Capo-chichi, C.D.; Cai, K.Q.; Simpkins, F.; Ganjei-Azar, P.; Godwin, A.K.; Xu, X.-X. Nuclear envelope structural defects cause chromosomal numerical instability and aneuploidy in ovarian cancer. BMC Med. 2011, 9, 28. [Google Scholar] [CrossRef]
  11. Evangelisti, C.; Rusciano, I.; Mongiorgi, S.; Ramazzotti, G.; Lattanzi, G.; Manzoli, L.; Cocco, L.; Ratti, S. The wide and growing range of lamin B-related diseases: From laminopathies to cancer. Cell. Mol. Life Sci. 2022, 79, 126. [Google Scholar] [CrossRef]
  12. Furukawa, K.; Inagaki, H.; Hotta, Y. Identification and cloning of an mRNA coding for a germ cell-specific A-type lamin in mice. Exp. Cell Res. 1994, 212, 426–430. [Google Scholar] [CrossRef]
  13. Furukawa, K.; Hotta, Y. cDNA cloning of a germ cell specific lamin B3 from mouse spermatocytes and analysis of its function by ectopic expression in somatic cells. EMBO J. 1993, 12, 97–106. [Google Scholar] [CrossRef]
  14. Shelton, K.R.; Egle, P.M.; Cochran, D.L. Nuclear envelope proteins: Identification of lamin B subtypes. Biochem. Biophys. Res. Commun. 1981, 103, 975–981. [Google Scholar] [CrossRef]
  15. Gruenbaum, Y.; Foisner, R. Lamins: Nuclear intermediate filament proteins with fundamental functions in nuclear mechanics and genome regulation. Annu. Rev. Biochem. 2015, 84, 131–164. [Google Scholar] [CrossRef]
  16. Heitlinger, E.; Peter, M.; Häner, M.; Lustig, A.; Aebi, U.; Nigg, E.A. Expression of chicken lamin B2 in Escherichia coli: Characterization of its structure, assembly, and molecular interactions. J. Cell Biol. 1991, 113, 485–495. [Google Scholar] [CrossRef]
  17. Shimi, T.; Pfleghaar, K.; Kojima, S.-i.; Pack, C.-G.; Solovei, I.; Goldman, A.E.; Adam, S.A.; Shumaker, D.K.; Kinjo, M.; Cremer, T.; et al. The A- and B-type nuclear lamin networks: Microdomains involved in chromatin organization and transcription. Genes Dev. 2008, 22, 3409–3421. [Google Scholar] [CrossRef]
  18. Shimi, T.; Kittisopikul, M.; Tran, J.; Goldman, A.E.; Adam, S.A.; Zheng, Y.; Jaqaman, K.; Goldman, R.D. Structural organization of nuclear lamins A, C, B1, and B2 revealed by superresolution microscopy. Mol. Biol. Cell 2015, 26, 4075–4086. [Google Scholar] [CrossRef]
  19. Machowska, M.; Piekarowicz, K.; Rzepecki, R. Regulation of lamin properties and functions: Does phosphorylation do it all? Open Biol. 2015, 5, 150094. [Google Scholar] [CrossRef]
  20. Peter, M.; Nakagawa, J.; Dorée, M.; Labbé, J.C.; Nigg, E.A. In vitro disassembly of the nuclear lamina and M phase-specific phosphorylation of lamins by cdc2 kinase. Cell 1990, 61, 591–602. [Google Scholar] [CrossRef]
  21. Hornbeck, P.; Huang, K.P.; Paul, W.E. Lamin B is rapidly phosphorylated in lymphocytes after activation of protein kinase C. Proc. Natl. Acad. Sci. USA 1988, 85, 2279–2283. [Google Scholar] [CrossRef]
  22. Obata, T.; Yaffe, M.B.; Leparc, G.G.; Piro, E.T.; Maegawa, H.; Kashiwagi, A.; Kikkawa, R.; Cantley, L.C. Peptide and protein library screening defines optimal substrate motifs for AKT/PKB. J. Biol. Chem. 2000, 275, 36108–36115. [Google Scholar] [CrossRef]
  23. Peter, M.; Sanghera, J.S.; Pelech, S.L.; Nigg, E.A. Mitogen-activated protein kinases phosphorylate nuclear lamins and display sequence specificity overlapping that of mitotic protein kinase p34cdc2. Eur. J. Biochem. 1992, 205, 287–294. [Google Scholar] [CrossRef]
  24. Li, H.; Roux, S.J. Casein kinase II protein kinase is bound to lamina-matrix and phosphorylates lamin-like protein in isolated pea nuclei. Proc. Natl. Acad. Sci. USA 1992, 89, 8434–8438. [Google Scholar] [CrossRef]
  25. Clements, L.; Manilal, S.; Love, D.R.; Morris, G.E. Direct interaction between emerin and lamin A. Biochem. Biophys. Res. Commun. 2000, 267, 709–714. [Google Scholar] [CrossRef]
  26. Ranade, D.; Pradhan, R.; Jayakrishnan, M.; Hegde, S.; Sengupta, K. Lamin A/C and Emerin depletion impacts chromatin organization and dynamics in the interphase nucleus. BMC Mol. Cell Biol. 2019, 20, 11. [Google Scholar] [CrossRef]
  27. Heald, R.; McKeon, F. Mutations of phosphorylation sites in lamin A that prevent nuclear lamina disassembly in mitosis. Cell 1990, 61, 579–589. [Google Scholar] [CrossRef]
  28. Shtivelman, E.; Sussman, J.; Stokoe, D. A role for PI 3-kinase and PKB activity in the G2/M phase of the cell cycle. Curr. Biol. 2002, 12, 919–924. [Google Scholar] [CrossRef]
  29. Cenni, V.; Bertacchini, J.; Beretti, F.; Lattanzi, G.; Bavelloni, A.; Riccio, M.; Ruzzene, M.; Marin, O.; Arrigoni, G.; Parnaik, V.; et al. Lamin A Ser404 is a nuclear target of Akt phosphorylation in C2C12 cells. J. Proteome Res. 2008, 7, 4727–4735. [Google Scholar] [CrossRef]
  30. Bertacchini, J.; Beretti, F.; Cenni, V.; Guida, M.; Gibellini, F.; Mediani, L.; Marin, O.; Maraldi, N.M.; de Pol, A.; Lattanzi, G.; et al. The protein kinase Akt/PKB regulates both prelamin A degradation and Lmna gene expression. FASEB J. 2013, 27, 2145–2155. [Google Scholar] [CrossRef]
  31. Naeem, A.S.; Zhu, Y.; Di, W.L.; Marmiroli, S.; O’Shaughnessy, R.F.L. AKT1-mediated Lamin A/C degradation is required for nuclear degradation and normal epidermal terminal differentiation. Cell Death Differ. 2015, 22, 2123–2132. [Google Scholar] [CrossRef]
  32. Jeong, S.; Ahn, J.; Jo, I.; Kang, S.-M.; Park, B.-J.; Cho, H.-S.; Kim, Y.-H.; Ha, N.-C. Cyclin-dependent kinase 1 depolymerizes nuclear lamin filaments by disrupting the head-to-tail interaction of the lamin central rod domain. J. Biol. Chem. 2022, 298, 102256. [Google Scholar] [CrossRef]
  33. Neumann-Staubitz, P.; Kitsberg, D.; Buxboim, A.; Neumann, H. A method to map the interaction network of the nuclear lamina with genetically encoded photo-crosslinkers in vivo. Front. Chem. 2022, 10, 905794. [Google Scholar] [CrossRef]
  34. Kochin, V.; Shimi, T.; Torvaldson, E.; Adam, S.A.; Goldman, A.; Pack, C.-G.; Melo-Cardenas, J.; Imanishi, S.Y.; Goldman, R.D.; Eriksson, J.E. Interphase phosphorylation of lamin A. J. Cell Sci. 2014, 127, 2683–2696. [Google Scholar] [CrossRef]
  35. Dephoure, N.; Zhou, C.; Villén, J.; Beausoleil, S.A.; Bakalarski, C.E.; Elledge, S.J.; Gygi, S.P. A quantitative atlas of mitotic phosphorylation. Proc. Natl. Acad. Sci. USA 2008, 105, 10762–10767. [Google Scholar] [CrossRef]
  36. Kutay, U.; Jühlen, R.; Antonin, W. Mitotic disassembly and reassembly of nuclear pore complexes. Trends Cell Biol. 2021, 31, 1019–1033. [Google Scholar] [CrossRef]
  37. Dubińska-Magiera, M.; Kozioł, K.; Machowska, M.; Piekarowicz, K.; Filipczak, D.; Rzepecki, R. Emerin Is Required for Proper Nucleus Reassembly after Mitosis: Implications for New Pathogenetic Mechanisms for Laminopathies Detected in EDMD1 Patients. Cells 2019, 8, 240. [Google Scholar] [CrossRef]
  38. Tsai, M.-Y.; Wang, S.; Heidinger, J.M.; Shumaker, D.K.; Adam, S.A.; Goldman, R.D.; Zheng, Y. A mitotic lamin B matrix induced by RanGTP required for spindle assembly. Science 2006, 311, 1887–1893. [Google Scholar] [CrossRef]
  39. Ungricht, R.; Kutay, U. Mechanisms and functions of nuclear envelope remodelling. Nat. Rev. Mol. Cell Biol. 2017, 18, 229–245. [Google Scholar] [CrossRef]
  40. McGowan, C.H.; Russell, P. The DNA damage response: Sensing and signaling. Curr. Opin. Cell Biol. 2004, 16, 629–633. [Google Scholar] [CrossRef]
  41. Roake, C.M.; Artandi, S.E. Regulation of human telomerase in homeostasis and disease. Nat. Rev. Mol. Cell Biol. 2020, 21, 384–397. [Google Scholar] [CrossRef]
  42. Barthel, F.P.; Wei, W.; Tang, M.; Martinez-Ledesma, E.; Hu, X.; Amin, S.B.; Akdemir, K.C.; Seth, S.; Song, X.; Wang, Q.; et al. Systematic analysis of telomere length and somatic alterations in 31 cancer types. Nat. Genet. 2017, 49, 349–357. [Google Scholar] [CrossRef]
  43. Crabbe, L.; Cesare, A.J.; Kasuboski, J.M.; Fitzpatrick, J.A.J.; Karlseder, J. Human telomeres are tethered to the nuclear envelope during post-mitotic nuclear assembly. Cell Rep. 2012, 2, 1521–1529. [Google Scholar] [CrossRef]
  44. Pennarun, G.; Picotto, J.; Etourneaud, L.; Redavid, A.-R.; Certain, A.; Gauthier, L.R.; Fontanilla-Ramirez, P.; Busso, D.; Chabance-Okumura, C.; Thézé, B.; et al. Increase in lamin B1 promotes telomere instability by disrupting the shelterin complex in human cells. Nucleic Acids Res. 2021, 49, 9886–9905. [Google Scholar] [CrossRef]
  45. Okamoto, K.; Bartocci, C.; Ouzounov, I.; Diedrich, J.K.; Yates, J.R.; Denchi, E.L. A two-step mechanism for TRF2-mediated chromosome end protection. Nature 2013, 494, 502–505. [Google Scholar] [CrossRef]
  46. Sarthy, J.; Bae, N.S.; Scrafford, J.; Baumann, P. Human RAP1 inhibits non-homologous end joining at telomeres. EMBO J. 2009, 28, 3390–3399. [Google Scholar] [CrossRef]
  47. van Steensel, B.; Smogorzewska, A.; de Lange, T. TRF2 protects human telomeres from end-to-end fusions. Cell 1998, 92, 401–413. [Google Scholar] [CrossRef]
  48. Mijit, M.; Caracciolo, V.; Melillo, A.; Amicarelli, F.; Giordano, A. Role of p53 in the Regulation of Cellular Senescence. Biomolecules 2020, 10, 420. [Google Scholar] [CrossRef]
  49. Qian, Y.; Chen, X. Senescence Regulation by the p53 Protein Family. Methods Mol. Biol. 2013, 965, 37–61. [Google Scholar] [CrossRef]
  50. Yoon, M.-H.; Kang, S.-M.; Lee, S.-J.; Woo, T.-G.; Oh, A.-Y.; Park, S.; Ha, N.-C.; Park, B.-J. p53 induces senescence through Lamin A/C stabilization-mediated nuclear deformation. Cell Death Dis. 2019, 10, 107. [Google Scholar] [CrossRef]
  51. Chuang, H.-H.; Wang, P.-H.; Niu, S.-W.; Zhen, Y.-Y.; Huang, M.-S.; Hsiao, M.; Yang, C.-J. Inhibition of FAK Signaling Elicits Lamin A/C-Associated Nuclear Deformity and Cellular Senescence. Front. Oncol. 2019, 9, 22. [Google Scholar] [CrossRef]
  52. Golubovskaya, V.M.; Finch, R.; Kweh, F.; Massoll, N.A.; Campbell-Thompson, M.; Wallace, M.R.; Cance, W.G. p53 regulates FAK expression in human tumor cells. Mol. Carcinog. 2008, 47, 373–382. [Google Scholar] [CrossRef]
  53. Goelzer, M.; Dudakovic, A.; Olcum, M.; Sen, B.; Ozcivici, E.; Rubin, J.; van Wijnen, A.J.; Uzer, G. Lamin A/C Is Dispensable to Mechanical Repression of Adipogenesis. Int. J. Mol. Sci. 2021, 22, 6580. [Google Scholar] [CrossRef]
  54. Panatta, E.; Butera, A.; Celardo, I.; Leist, M.; Melino, G.; Amelio, I. p53 regulates expression of nuclear envelope components in cancer cells. Biol. Direct 2022, 17, 38. [Google Scholar] [CrossRef]
  55. Lämmerhirt, L.; Kappelmann-Fenzl, M.; Fischer, S.; Pommer, M.; Zimmermann, T.; Kluge, V.; Matthies, A.; Kuphal, S.; Bosserhoff, A.K. Knockdown of Lamin B1 and the Corresponding Lamin B Receptor Leads to Changes in Heterochromatin State and Senescence Induction in Malignant Melanoma. Cells 2022, 11, 2154. [Google Scholar] [CrossRef]
  56. Freund, A.; Laberge, R.-M.; Demaria, M.; Campisi, J. Lamin B1 loss is a senescence-associated biomarker. Mol. Biol. Cell 2012, 23, 2066–2075. [Google Scholar] [CrossRef]
  57. Onorati, A.; Havas, A.P.; Lin, B.; Rajagopal, J.; Sen, P.; Adams, P.D.; Dou, Z. Upregulation of PD-L1 in Senescence and Aging. Mol. Cell. Biol. 2022, 42, e0017122. [Google Scholar] [CrossRef]
  58. Iwabuchi, K.; Bartel, P.L.; Li, B.; Marraccino, R.; Fields, S. Two cellular proteins that bind to wild-type but not mutant p53. Proc. Natl. Acad. Sci. USA 1994, 91, 6098–6102. [Google Scholar] [CrossRef]
  59. Bothmer, A.; Robbiani, D.F.; Feldhahn, N.; Gazumyan, A.; Nussenzweig, A.; Nussenzweig, M.C. 53BP1 regulates DNA resection and the choice between classical and alternative end joining during class switch recombination. J. Exp. Med. 2010, 207, 855–865. [Google Scholar] [CrossRef]
  60. Botuyan, M.V.; Lee, J.; Ward, I.M.; Kim, J.-E.; Thompson, J.R.; Chen, J.; Mer, G. Structural Basis for the Methylation State-Specific Recognition of Histone H4-K20 by 53BP1 and Crb2 in DNA Repair. Cell 2006, 127, 1361–1373. [Google Scholar] [CrossRef]
  61. Fradet-Turcotte, A.; Canny, M.D.; Escribano-Díaz, C.; Orthwein, A.; Leung, C.C.Y.; Huang, H.; Landry, M.-C.; Kitevski-LeBlanc, J.; Noordermeer, S.M.; Sicheri, F.; et al. 53BP1 is a reader of the DNA-damage-induced H2A Lys 15 ubiquitin mark. Nature 2013, 499, 50–54. [Google Scholar] [CrossRef]
  62. Gibbs-Seymour, I.; Markiewicz, E.; Bekker-Jensen, S.; Mailand, N.; Hutchison, C.J. Lamin A/C-dependent interaction with 53BP1 promotes cellular responses to DNA damage. Aging Cell 2015, 14, 162–169. [Google Scholar] [CrossRef]
  63. Gonzalez-Suarez, I.; Redwood, A.B.; Perkins, S.M.; Vermolen, B.; Lichtensztejin, D.; Grotsky, D.A.; Morgado-Palacin, L.; Gapud, E.J.; Sleckman, B.P.; Sullivan, T.; et al. Novel roles for A-type lamins in telomere biology and the DNA damage response pathway. EMBO J. 2009, 28, 2414–2427. [Google Scholar] [CrossRef]
  64. Etourneaud, L.; Moussa, A.; Rass, E.; Genet, D.; Willaume, S.; Chabance-Okumura, C.; Wanschoor, P.; Picotto, J.; Thézé, B.; Dépagne, J.; et al. Lamin B1 sequesters 53BP1 to control its recruitment to DNA damage. Sci. Adv. 2021, 7, eabb3799. [Google Scholar] [CrossRef]
  65. Chen, S.N.; Lombardi, R.; Karmouch, J.; Tsai, J.-Y.; Czernuszewicz, G.; Taylor, M.R.G.; Mestroni, L.; Coarfa, C.; Gurha, P.; Marian, A.J. DNA Damage Response/TP53 Pathway Is Activated and Contributes to the Pathogenesis of Dilated Cardiomyopathy Associated With LMNA (Lamin A/C) Mutations. Circ. Res. 2019, 124, 856–873. [Google Scholar] [CrossRef]
  66. Wang, J.; Kondo, T.; Nakazawa, T.; Oishi, N.; Mochizuki, K.; Katoh, R. Constitutional abnormality of nuclear membrane proteins in small cell lung carcinoma. Virchows Arch. 2019, 475, 407–414. [Google Scholar] [CrossRef]
  67. Jia, Y.; Vong, J.S.-L.; Asafova, A.; Garvalov, B.K.; Caputo, L.; Cordero, J.; Singh, A.; Boettger, T.; Günther, S.; Fink, L.; et al. Lamin B1 loss promotes lung cancer development and metastasis by epigenetic derepression of RET. J. Exp. Med. 2019, 216, 1377–1395. [Google Scholar] [CrossRef]
  68. Stefanello, S.T.; Luchtefeld, I.; Liashkovich, I.; Pethö, Z.; Azzam, I.; Bulk, E.; Rosso, G.; Döhlinger, L.; Hesse, B.; Oeckinghaus, A.; et al. Impact of the Nuclear Envelope on Malignant Transformation, Motility, and Survival of Lung Cancer Cells. Adv. Sci. 2021, 8, e2102757. [Google Scholar] [CrossRef]
  69. Rubporn, A.; Srisomsap, C.; Subhasitanont, P.; Chokchaichamnankit, D.; Chiablaem, K.; Svasti, J.; Sangvanich, P. Comparative proteomic analysis of lung cancer cell line and lung fibroblast cell line. Cancer Genom. Proteom. 2009, 6, 229–237. [Google Scholar]
  70. Hu, C.; Zhou, A.; Hu, X.; Xiang, Y.; Huang, M.; Huang, J.; Yang, D.; Tang, Y. LMNA Reduced Acquired Resistance to Erlotinib in NSCLC by Reversing the Epithelial-Mesenchymal Transition via the FGFR/MAPK/c-fos Signaling Pathway. Int. J. Mol. Sci. 2022, 23, 13237. [Google Scholar] [CrossRef]
  71. Li, W.; Li, X.; Li, X.; Li, M.; Yang, P.; Wang, X.; Li, L.; Yang, B. Lamin B1 Overexpresses in Lung Adenocarcinoma and Promotes Proliferation in Lung Cancer Cells via AKT Pathway. Onco. Targets. Ther. 2020, 13, 3129–3139. [Google Scholar] [CrossRef]
  72. Li, J.; Sun, Z.; Cui, Y.; Qin, L.; Wu, F.; Li, Y.; Du, N.; Li, X. Knockdown of LMNB1 Inhibits the Proliferation of Lung Adenocarcinoma Cells by Inducing DNA Damage and Cell Senescence. Front. Oncol. 2022, 12, 913740. [Google Scholar] [CrossRef]
  73. Jiao, X.; Gao, W.; Ren, H.; Wu, Y.; Li, T.; Li, S.; Yan, H. Kruppel like factor 16 promotes lung adenocarcinoma progression by upregulating lamin B2. Bioengineered 2022, 13, 9482–9494. [Google Scholar] [CrossRef]
  74. Ma, Y.; Fei, L.; Zhang, M.; Zhang, W.; Liu, X.; Wang, C.; Luo, Y.; Zhang, H.; Han, Y. Lamin B2 binding to minichromosome maintenance complex component 7 promotes non-small cell lung carcinogenesis. Oncotarget 2017, 8, 104813–104830. [Google Scholar] [CrossRef]
  75. Zhang, M.-Y.; Han, Y.-C.; Han, Q.; Liang, Y.; Luo, Y.; Wei, L.; Yan, T.; Yang, Y.; Liu, S.-L.; Wang, E.-H. Lamin B2 promotes the malignant phenotype of non-small cell lung cancer cells by upregulating dimethylation of histone 3 lysine 9. Exp. Cell Res. 2020, 393, 112090. [Google Scholar] [CrossRef]
  76. Hatch, E.M.; Fischer, A.H.; Deerinck, T.J.; Hetzer, M.W. Catastrophic nuclear envelope collapse in cancer cell micronuclei. Cell 2013, 154, 47–60. [Google Scholar] [CrossRef]
  77. de Leeuw, R.; Gruenbaum, Y.; Medalia, O. Nuclear Lamins: Thin Filaments with Major Functions. Trends Cell Biol. 2018, 28, 34–45. [Google Scholar] [CrossRef]
  78. White, D.E.; Negorev, D.; Peng, H.; Ivanov, A.V.; Maul, G.G.; Rauscher, F.J. KAP1, a novel substrate for PIKK family members, colocalizes with numerous damage response factors at DNA lesions. Cancer Res. 2006, 66, 11594–11599. [Google Scholar] [CrossRef]
  79. Sanchez, R.; Zhou, M.-M. The PHD finger: A versatile epigenome reader. Trends Biochem. Sci. 2011, 36, 364–372. [Google Scholar] [CrossRef]
  80. Filippakopoulos, P.; Knapp, S. The bromodomain interaction module. FEBS Lett. 2012, 586, 2692–2704. [Google Scholar] [CrossRef]
  81. Wang, C.; Ivanov, A.; Chen, L.; Fredericks, W.J.; Seto, E.; Rauscher, F.J.; Chen, J. MDM2 interaction with nuclear corepressor KAP1 contributes to p53 inactivation. EMBO J. 2005, 24, 3279–3290. [Google Scholar] [CrossRef]
  82. Wang, C.; Rauscher, F.J.; Cress, W.D.; Chen, J. Regulation of E2F1 function by the nuclear corepressor KAP1. J. Biol. Chem. 2007, 282, 29902–29909. [Google Scholar] [CrossRef]
  83. Gesson, K.; Rescheneder, P.; Skoruppa, M.P.; von Haeseler, A.; Dechat, T.; Foisner, R. A-type lamins bind both hetero- and euchromatin, the latter being regulated by lamina-associated polypeptide 2 alpha. Genome Res. 2016, 26, 462–473. [Google Scholar] [CrossRef]
  84. Chuang, H.-H.; Huang, M.-S.; Zhen, Y.-Y.; Chuang, C.-H.; Lee, Y.-R.; Hsiao, M.; Yang, C.-J. FAK Executes Anti-Senescence via Regulating EZH2 Signaling in Non-Small Cell Lung Cancer Cells. Biomedicines 2022, 10, 1937. [Google Scholar] [CrossRef]
  85. Cao, R.; Wang, L.; Wang, H.; Xia, L.; Erdjument-Bromage, H.; Tempst, P.; Jones, R.S.; Zhang, Y. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science 2002, 298, 1039–1043. [Google Scholar] [CrossRef]
  86. Duan, R.; Du, W.; Guo, W. EZH2: A novel target for cancer treatment. J. Hematol. Oncol. 2020, 13, 104. [Google Scholar] [CrossRef]
  87. Yu, Y.; Qi, J.; Xiong, J.; Jiang, L.; Cui, D.; He, J.; Chen, P.; Li, L.; Wu, C.; Ma, T.; et al. Epigenetic Co-Deregulation of EZH2/TET1 is a Senescence-Countering, Actionable Vulnerability in Triple-Negative Breast Cancer. Theranostics 2019, 9, 761–777. [Google Scholar] [CrossRef]
  88. Chibaya, L.; Murphy, K.C.; DeMarco, K.D.; Gopalan, S.; Liu, H.; Parikh, C.N.; Lopez-Diaz, Y.; Faulkner, M.; Li, J.; Morris, J.P.; et al. EZH2 inhibition remodels the inflammatory senescence-associated secretory phenotype to potentiate pancreatic cancer immune surveillance. Nat. Cancer 2023, 4, 872–892. [Google Scholar] [CrossRef]
  89. Mulligan, L.M. RET revisited: Expanding the oncogenic portfolio. Nat. Rev. Cancer 2014, 14, 173–186. [Google Scholar] [CrossRef]
  90. Otsuki, Y.; Saya, H.; Arima, Y. Prospects for new lung cancer treatments that target EMT signaling. Dev. Dyn. 2018, 247, 462–472. [Google Scholar] [CrossRef]
  91. Pascual-Reguant, L.; Blanco, E.; Galan, S.; Le Dily, F.; Cuartero, Y.; Serra-Bardenys, G.; Di Carlo, V.; Iturbide, A.; Cebrià-Costa, J.P.; Nonell, L.; et al. Lamin B1 mapping reveals the existence of dynamic and functional euchromatin lamin B1 domains. Nat. Commun. 2018, 9, 3420. [Google Scholar] [CrossRef]
  92. Zhao, C.-C.; Chen, J.; Zhang, L.-Y.; Liu, H.; Zhang, C.-G.; Liu, Y. Lamin B2 promotes the progression of triple negative breast cancer via mediating cell proliferation and apoptosis. Biosci. Rep. 2021, 41, BSR20203874. [Google Scholar] [CrossRef]
  93. Fracchia, A.; Asraf, T.; Salmon-Divon, M.; Gerlitz, G. Increased Lamin B1 Levels Promote Cell Migration by Altering Perinuclear Actin Organization. Cells 2020, 9, 2161. [Google Scholar] [CrossRef]
  94. Dong, C.-H.; Jiang, T.; Yin, H.; Song, H.; Zhang, Y.; Geng, H.; Shi, P.-C.; Xu, Y.-X.; Gao, H.; Liu, L.-Y.; et al. LMNB2 promotes the progression of colorectal cancer by silencing p21 expression. Cell Death Dis. 2021, 12, 331. [Google Scholar] [CrossRef]
  95. Belt, E.J.T.; Fijneman, R.J.A.; van den Berg, E.G.; Bril, H.; Delis-van Diemen, P.M.; Tijssen, M.; van Essen, H.F.; de Lange-de Klerk, E.S.M.; Beliën, J.A.M.; Stockmann, H.B.A.C.; et al. Loss of lamin A/C expression in stage II and III colon cancer is associated with disease recurrence. Eur. J. Cancer 2011, 47, 1837–1845. [Google Scholar] [CrossRef]
  96. Moss, S.; Krivosheyev, V.; de Souza, A.; Chin, K.; Gaetz, H.; Chaudhary, N.; Worman, H.; Holt, P. Decreased and aberrant nuclear lamin expression in gastrointestinal tract neoplasms. Gut 1999, 45, 723–729. [Google Scholar] [CrossRef]
  97. Willis, N.D.; Cox, T.R.; Rahman-Casañs, S.F.; Smits, K.; Przyborski, S.A.; van den Brandt, P.; van Engeland, M.; Weijenberg, M.; Wilson, R.G.; de Bruïne, A.; et al. Lamin A/C Is a Risk Biomarker in Colorectal Cancer. PLoS ONE 2008, 3, e2988. [Google Scholar] [CrossRef]
  98. Wu, Z.; Wu, L.; Weng, D.; Xu, D.; Geng, J.; Zhao, F. Reduced expression of lamin A/C correlates with poor histological differentiation and prognosis in primary gastric carcinoma. J. Exp. Clin. Cancer Res. 2009, 28, 8. [Google Scholar] [CrossRef]
  99. Roncato, F.; Regev, O.; Feigelson, S.W.; Yadav, S.K.; Kaczmarczyk, L.; Levi, N.; Drago-Garcia, D.; Ovadia, S.; Kizner, M.; Addadi, Y.; et al. Reduced Lamin A/C Does Not Facilitate Cancer Cell Transendothelial Migration but Compromises Lung Metastasis. Cancers 2021, 13, 2383. [Google Scholar] [CrossRef]
  100. Nishikawa, T.; Kuwano, Y.; Nakata, M.; Rokutan, K.; Nishida, K. Multiple G-quadruplexes in the LMNA promoter regulate LMNA variant 6 transcription and promote colon cancer cell growth. Biochim. Biophys. Acta Gene Regul. Mech. 2021, 1864, 194746. [Google Scholar] [CrossRef]
  101. Sengupta, D.; Sengupta, K. Elevated levels of lamin A promote HR and NHEJ-mediated repair mechanisms in etoposide-treated ovarian cancer cells. bioRxiv 2022. [Google Scholar] [CrossRef]
  102. Urciuoli, E.; D’Oria, V.; Petrini, S.; Peruzzi, B. Lamin A/C Mechanosensor Drives Tumor Cell Aggressiveness and Adhesion on Substrates With Tissue-Specific Elasticity. Front. Cell Dev. Biol. 2021, 9, 712377. [Google Scholar] [CrossRef]
  103. Skvortsov, S.; Schäfer, G.; Stasyk, T.; Fuchsberger, C.; Bonn, G.K.; Bartsch, G.; Klocker, H.; Huber, L.A. Proteomics profiling of microdissected low- and high-grade prostate tumors identifies Lamin A as a discriminatory biomarker. J. Proteome Res. 2011, 10, 259–268. [Google Scholar] [CrossRef]
  104. Kong, L.; Schäfer, G.; Bu, H.; Zhang, Y.; Zhang, Y.; Klocker, H. Lamin A/C protein is overexpressed in tissue-invading prostate cancer and promotes prostate cancer cell growth, migration and invasion through the PI3K/AKT/PTEN pathway. Carcinogenesis 2012, 33, 751–759. [Google Scholar] [CrossRef]
  105. Gatti, G.; Vilardo, L.; Musa, C.; Di Pietro, C.; Bonaventura, F.; Scavizzi, F.; Torcinaro, A.; Bucci, B.; Saporito, R.; Arisi, I.; et al. Role of Lamin A/C as Candidate Biomarker of Aggressiveness and Tumorigenicity in Glioblastoma Multiforme. Biomedicines 2021, 9, 1343. [Google Scholar] [CrossRef]
  106. Chiarini, F.; Paganelli, F.; Balestra, T.; Capanni, C.; Fazio, A.; Manara, M.C.; Landuzzi, L.; Petrini, S.; Evangelisti, C.; Lollini, P.-L.; et al. Lamin A and the LINC complex act as potential tumor suppressors in Ewing Sarcoma. Cell Death Dis. 2022, 13, 346. [Google Scholar] [CrossRef]
  107. Sun, S.; Xu, M.Z.; Poon, R.T.; Day, P.J.; Luk, J.M. Circulating Lamin B1 (LMNB1) biomarker detects early stages of liver cancer in patients. J. Proteome Res. 2010, 9, 70–78. [Google Scholar] [CrossRef]
  108. Hong, J.-H.; Liang, S.-T.; Wang, A.S.-S.; Yeh, C.-M.; Huang, H.-P.; Sun, C.-D.; Zhang, Z.-H.; Lu, S.-Y.; Chao, Y.-H.; Chen, C.-H.; et al. LMNB1, a potential marker for early prostate cancer progression. Am. J. Cancer Res. 2022, 12, 3390–3404. [Google Scholar]
  109. Li, L.; Du, Y.; Kong, X.; Li, Z.; Jia, Z.; Cui, J.; Gao, J.; Wang, G.; Xie, K. Lamin B1 Is a Novel Therapeutic Target of Betulinic Acid in Pancreatic Cancer. Clin. Cancer Res. 2013, 19, 4651–4661. [Google Scholar] [CrossRef]
  110. Bengtsson, S.; Krogh, M.; Szigyarto, C.A.-K.; Uhlen, M.; Schedvins, K.; Silfverswärd, C.; Linder, S.; Auer, G.; Alaiya, A.; James, P. Large-scale proteomics analysis of human ovarian cancer for biomarkers. J. Proteome Res. 2007, 6, 1440–1450. [Google Scholar] [CrossRef]
  111. Yang, Y.; Gao, L.; Chen, J.; Xiao, W.; Liu, R.; Kan, H. Lamin B1 is a potential therapeutic target and prognostic biomarker for hepatocellular carcinoma. Bioengineered 2022, 13, 9211–9231. [Google Scholar] [CrossRef]
  112. Radspieler, M.M.; Schindeldecker, M.; Stenzel, P.; Försch, S.; Tagscherer, K.E.; Herpel, E.; Hohenfellner, M.; Hatiboglu, G.; Roth, W.; Macher-Goeppinger, S. Lamin-B1 is a senescence-associated biomarker in clear-cell renal cell carcinoma. Oncol. Lett. 2019, 18, 2654–2660. [Google Scholar] [CrossRef]
Figure 1. Lamin family members and prominent phosphorylation sites. (A) Schematic summary of the structure common to all lamin subtypes. (B) Overview of known lamin splice variants encoded by LMNA (blue), LMNB1 (brown), and LMNB2 (orange). (C) Major lamin phosphorylation sites targeted by oncogenes including CDK1 and AKT (PKB).
Figure 1. Lamin family members and prominent phosphorylation sites. (A) Schematic summary of the structure common to all lamin subtypes. (B) Overview of known lamin splice variants encoded by LMNA (blue), LMNB1 (brown), and LMNB2 (orange). (C) Major lamin phosphorylation sites targeted by oncogenes including CDK1 and AKT (PKB).
Cancers 15 05501 g001
Figure 2. The multifaceted roles of lamins (orange, green) in maintaining cellular integrity and the DNA damage response in physiological conditions (left) and altered functions in lung cancer (right). Major regulators of cell cycle (Cyclins D and E, p53, 53BP1), proliferation (AKT), and chromatin accessibility (EZH2) interact with the lamin network to maintain cellular homeostasis and to coordinate structural integrity before and during cell division. In cancer, hijacking of these functions either by the mutational activation or the inactivation of proto-oncogenes and tumor suppressor genes contributes to the modulation and reprogramming of the lamin network, respectively. Specifically, aberrant AKT and cyclin D signaling in lung cancer induces B-type lamin depolymerization and enhanced proliferation, while the EZH2-mediated regulation of lamin A/C alters the transcriptional control of factors promoting cancer aggressiveness and EMT by down-regulating E-cadherin and upregulating vimentin. The arrows show up- and downregulation of the indicated factors altered by changes in lamin levels.
Figure 2. The multifaceted roles of lamins (orange, green) in maintaining cellular integrity and the DNA damage response in physiological conditions (left) and altered functions in lung cancer (right). Major regulators of cell cycle (Cyclins D and E, p53, 53BP1), proliferation (AKT), and chromatin accessibility (EZH2) interact with the lamin network to maintain cellular homeostasis and to coordinate structural integrity before and during cell division. In cancer, hijacking of these functions either by the mutational activation or the inactivation of proto-oncogenes and tumor suppressor genes contributes to the modulation and reprogramming of the lamin network, respectively. Specifically, aberrant AKT and cyclin D signaling in lung cancer induces B-type lamin depolymerization and enhanced proliferation, while the EZH2-mediated regulation of lamin A/C alters the transcriptional control of factors promoting cancer aggressiveness and EMT by down-regulating E-cadherin and upregulating vimentin. The arrows show up- and downregulation of the indicated factors altered by changes in lamin levels.
Cancers 15 05501 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Janetzko, J.; Oeck, S.; Schramm, A. The Multifaceted Roles of Lamins in Lung Cancer and DNA Damage Response. Cancers 2023, 15, 5501. https://doi.org/10.3390/cancers15235501

AMA Style

Janetzko J, Oeck S, Schramm A. The Multifaceted Roles of Lamins in Lung Cancer and DNA Damage Response. Cancers. 2023; 15(23):5501. https://doi.org/10.3390/cancers15235501

Chicago/Turabian Style

Janetzko, Janina, Sebastian Oeck, and Alexander Schramm. 2023. "The Multifaceted Roles of Lamins in Lung Cancer and DNA Damage Response" Cancers 15, no. 23: 5501. https://doi.org/10.3390/cancers15235501

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop