Figure 8 - uploaded by Emily Helene Sage
Content may be subject to copyright.
Immunolocalization of hevin in normal mouse cerebellum. IHC analysis of methyl Carnoy’s-fixed, paraffin-embedded sections of mouse brain with MAb 12-155 (red). Two ϫ 200 magnification fields ( A,B ) and two ϫ 400 magnification fields ( C,D ) are shown. Reactivity was visualized after incubation with Cy3-conju- gated anti-rat IgG secondary antibody. The slides were counterstained with Hoechst 22258 (blue). MAb TUJ1, specific for neuronal ␤ -actin, was used to identify neurons and was visualized after incubation with FITC- conjugated anti-mouse IgG (green). Arrows in C indicate reactivity with Bergmann glia and/or Purkinje cells in the molecular layer. pl, Purkinje layer; gl, granular layer; ml, molecular layer; w, white matter. 

Immunolocalization of hevin in normal mouse cerebellum. IHC analysis of methyl Carnoy’s-fixed, paraffin-embedded sections of mouse brain with MAb 12-155 (red). Two ϫ 200 magnification fields ( A,B ) and two ϫ 400 magnification fields ( C,D ) are shown. Reactivity was visualized after incubation with Cy3-conju- gated anti-rat IgG secondary antibody. The slides were counterstained with Hoechst 22258 (blue). MAb TUJ1, specific for neuronal ␤ -actin, was used to identify neurons and was visualized after incubation with FITC- conjugated anti-mouse IgG (green). Arrows in C indicate reactivity with Bergmann glia and/or Purkinje cells in the molecular layer. pl, Purkinje layer; gl, granular layer; ml, molecular layer; w, white matter. 

Source publication
Article
Full-text available
Hevin, also known as SC1, MAST 9, SPARC-like 1, RAGS1 and ECM2, is a member of the SPARC-related family of matricellular proteins. Mouse hevin is 53% identical to mouse SPARC, and both proteins share a follistatin-like module and an extracellular Ca(2+)-binding (E-C) domain. SPARC functions as a modulator of cell-matrix interactions, a regulator of...

Context in source publication

Context 1
... heterogeneity in posttranslational modification of the full-length protein. Recoveries of recombinant hevin are shown in Table 1. Based on scanning densitometry of SDS-polyacrylamide gels, which tends to overestimate the con- tribution of impurities, preparations of recombinant hevin were ف 80% pure after elution from an isoelectric focusing column. Greater yields, however, with less purity, were obtained from size-exclusion chromatography compared with isoelectric focusing. Figures 3 and 4 confirm that recombinant hevin (rhevin), expressed and purified as described above, is biologi- cally active. Human hevin has been described as antiadhesive for cultured human umbilical vein endothelial cells (HUVECs) (Girard and Springer 1996). We have confirmed these data and have extended the findings to define an anti-adhesive activity of mouse recombinant hevin on BAECs in vitro. As shown in Figures 3A and 3B, purified hevin, added at 10 ␮ g/ml to cells in suspension, inhibited the spreading of BAECs on tissue culture plastic (i.e., was anti-adhesive) in the presence of 0% FBS. This effect was apparent up to 6 hr after plating, after which hevin-treated BAECs completed adhesion and resembled non-treated or BSA-treated controls. Quantification of these data as Rounding Indices is shown in Figure 4. These results were also obtained in the presence of 1% FBS (not shown). We also measured the anti-adhesive activity of recombinant hevin presented as a substrate to trypsin- released BAECs. As shown in Figure 5, BAECs in the absence of FBS adhered to substrates coated with fibronectin (Figures 5E and 5H), but were rounded and clumped on hevin-coated wells (Figures 5F and 5I). This effect was maintained over ف 12 hr, after which most of the cells began to spread. Intermediate levels of adhesion that were time-dependent were seen with substrates coated with 10% FBS, and rounding similar to that shown in Figures 5C, 5F, 5I, and 5L was observed with substrates coated with recombinant SPARC (not shown). Cell viability was maintained under all conditions. Partially purified hevin was used to immunize rats to produce MAbs specific for mouse hevin. This protocol resulted in over 200 hybridoma clones that bound strongly to hevin by ELISA (data not shown). These reagents were acutely needed because there were no anti-hevin antibodies that reacted reliably with mouse tissues or with paraformaldehyde-fixed tissue, that blocked any activity of native hevin, or that could immunoprecipitate native antigen. For selection of hybridomas, tissue culture supernatants from the clones were screened for reactivity with recombinant hevin in a variety of assays. A partial list of the most ex- tensively characterized rat anti-hevin hybridomas is shown in Table 2. The majority of MAbs are IgG 2a , and each clone reacts with mouse hevin by indirect ELISA. In contrast, many MAbs failed to react with hevin in solution, as determined by a capture ELISA (Table 2). Several of the MAbs described in Table 2 were used to identify hevin in mouse brain lysates (Figure 6A, Lane 1). In each lysate, a band of M r 105,000–110,000 was the predominant immunoreactive species detected by three different MAbs. Minor bands most likely re- flect differences in posttranslational modification of the hevin chain (Hambrock et al. 2003) because tissue lysates contain secreted as well as cellular (endoplas- mic reticulum and Golgi-associated) hevin, with dif- fering degrees of glycosylation. Immunoprecipitation of hevin from extracts of mouse brain by MAb 12-18 also showed a protein of M r 105,000–110,000 by SDS-PAGE (Table 2; Figure 6B, Lane 2). In no cases was immunoreactivity obtained with proteins in lysates from hevin-null brains (Figure 6A, Lanes 2). The disparity in M r of hevin observed in Figures 1 and 2 vs Figure 6 is likely due to the differences in polyacrylamide gel composition (10% in Figures 1 and 2 vs 4–12% gradient in Figure 6), as well as in buffer conditions (Tris-HC1 vs Bis-Tris). We found that MAb 12-51 was particularly useful for IHC localization of hevin in frozen or paraffin-embedded tissue because it also crossreacts with human hevin. Figure 7 shows the reactivity of 12-51 with human skin and mouse kidney. Strong reactivity was apparent in the dermal layers of the skin, as well as in muscle and the epithelia of glands (Figures 7A and 7B). In the brain, 12-51 highlighted specific cells in the parenchyma of the cerebellum and also vessels in other regions (not shown). Arterioles, venules, and capillaries in the dermis were also reactive with MAb 12-51 (Figure 7B). In paraffin-embedded mouse kidney, hevin was localized by MAb 12-155 to certain glomeruli and tubule epithelial cells (Figure 7C). These data are consistent with previous results in which hevin mRNA was localized to vessels, epidermis, and selected populations of renal tubules by ISH (Soderling et al. 1997). Staining for hevin in mouse cerebellum with MAb 12-155 revealed patterns as shown in Figure 8. Hevin, shown in red, localized conspicuously to the Purkinje cell layer (Figures 8A and 8B) and to the Bergmann glia and Purkinje cells throughout the molecular layer (Figures 8C and 8D). Neurons, shown in green, were localized with MAb TUJ1, which is specific for neuronal class III ␤ -tubulin; nuclei, shown in blue, were stained with Hoechst 22258. These results extend ear- lier reports of hevin mRNA expression in the Purkinje cell layer (McKinnon and Margolskee 1996; Sullivan and Sage 2004). We also evaluated the production of hevin in a limited panel of cancerous tissues (Figure 9). Of the tumor tissues that we screened (human colorectal, lung, breast, pancreatic, and endometrial adenocarcinoma), reactivity was apparent only in endometrial carcinoma associated with the luminal duct epithelial layer (Figure 9G). Although there was no reactivity in two samples of human pancreatic adenocarcinoma, we found tumor as well as stromal reactivity with MAbs 12-51 and 12-155 in human pancreatic tumor xenografts grown in immunocompromised mice (not shown). Xe- nografts of human non-small-cell lung carcinoma from immunocompromised mice are shown in Figures 9A–9F), after incubation with several additional anti- hevin MAbs (identified on each panel) that have not been listed in Table 2. Each of these staining patterns reflects a stromal/vascular component that was identified by the respective anti-hevin MAb. These data pro- vide evidence for hevin as a stroma-associated protein of neoplastic tissue, an observation consistent with findings by other investigators from serial analysis of gene expression (SAGE) analysis of invasive pancreatic carcinomas (Ryu et al. 2001; Iacobuzio-Donahue et al. 2003). Matricellular proteins regulate the interaction of cells with the ECM, a critical function in the maintenance of tissue homeostasis. Hevin, originally described in rodents as SC-1, is a matricellular protein belonging to the SPARC family. In the present study we have produced recombinant mouse hevin and have purified it to near homogeneity, developed MAbs specific for hevin, and identified a function of hevin in vitro. The MAbs described herein will be useful for elucidating the functions of hevin in vivo. For example, hevin has recently been shown to be identical to a neuronal guidance protein, the antigen for the MAb RAGS1 (Gongidi et al. 2004). RAGS1 (Radial Glial Stop Sig- nal molecule 1) recognizes an antigen on the surface of radial glial cells that is distributed where neurons ter- minate their migration. Gongidi et al. (2004) found that the MAbs RAGS1 and 12-155 react on immuno- blots with the same protein. Moreover, inhibition of hevin with RAGS1 allowed neurons to migrate past the normal termination point in the cerebral cortex, thereby demonstrating that hevin is critical for appropriate neuron migration. Among the many cell lines that have been tested for the production of hevin, the few that produce hevin at detectable levels by immunoblotting are tumor lines. Lysates of hevin-producing tumor cells yield a primary immunoreactive band of M r 130,000 on SDS- PAGE using 10% polyacrylamide gels (not shown). We found hevin of similar M r in lysates of human and mouse brain tissue, or of M r 105,000 on 4–12% Bis- Tris gradient gels. The major immunoreactive band of recombinant mouse hevin migrates slightly faster than the M r 130,000 band characteristic of cell and tissue lysates, a possible consequence of the inability of insect cells to produce N -glycoproteins containing a sialic acid cap, a modification of many complex carbohydrate chains of secreted proteins synthesized by mammalian cells. Note that the apparent M r of recombinant hevin is 105,000 when estimated by separation on a NuPAGE 4–12% Bis-Tris gel with NuPAGE MOPS SDS running buffer (Invitrogen) (Figure 6). Estimates of the M r of human hevin in the liter- ature vary from 95,000 (Hambrock et al. 2003) to 150,000 (Bendik et al. 1998) by SDS-PAGE. Therefore, we consider acceptable the apparent size varia- tion that we have observed in recombinant murine hevin and explain it as a possible consequence of the gel resolution system and/or interaction of the acidic N-terminal domain with buffers of varying pH and/or ionic strength. The de-adhesive effects of hevin that we report are consistent with a prior study. Girard and Springer (1996) found that hevin (in contrast to fibronectin, thrombospondin 1, and tenascin C) was non-permis- sive for attachment and spreading by HUVECs and BAECs. Furthermore, it diminished adhesion to fibronectin, an effect correlated with reduced formation of focal adhesions. Initially, it was suggested that hevin supported lymphocyte extravasion through high endothelial venules via the diminishment of cell–cell and/or cell–ECM contacts in a dynamic, chemokine- responsive manner (Girard and Springer 1995,1996). We speculate that hevin might play a comparable role in the desmoplastic endothelium associated with tumor ...

Citations

... SPARCL1 is an antiadhesive glycoprotein and functions as the extracellular protein. [12][13][14] It can regulate cell-matrix interactions and participates in tissue remodeling. [25][26][27] Compelling evidence has confirmed that matricellular proteins are the important regulators of neuronal integrity. ...
Article
Full-text available
Background Secreted protein acidic and rich in cysteine-like 1 (SPARCL1) regulates synaptic stability and is up-regulated during axonal regeneration. Here, serum SPARCL1 was determined for estimating severity and prognosticating early neurological deterioration (END) and functional outcomes of acute intracerebral hemorrhage (ICH). Methods In this prospective observational cohort study of 156 patients with supratentorial ICH, blood samples of 53 were acquired not only at admission but also ad days 1, 3, 5, 7 and 10. Another group of 53 healthy controls were recruited. The modified Rankin Scale (mRS) scores of 3–6 at poststroke six months were regarded as poor prognosis. Results As opposed to controls, serum SPARCL1 levels were markedly elevated during the early ten days after ICH, with the highest levels at days 1 and 3. Admission serum SPARCL1 levels were independently correlated with National Institutes of Health Stroke Scale scores and hematoma volume, were significantly increased in the order of six-month mRS scores from 0 to 6 and were independently correlated with six-month mRS scores. Serum SPARCL1 levels were linearly related to risks of poor six-month prognosis and END under restricted cubic spline, had significant efficiency under receiver operating characteristic (ROC) curve and were independently associated with END and poor prognosis. Subgroup analysis confirmed that no interactions existed for associations of serum SPARCL1 levels with other variables, such as age, gender and some specific vascular risk factors. END and poor prognosis prediction models integrating serum SPARCL1 were displayed using the two nomograms. The poor prognosis prediction model, but END prediction model not, performed well under calibration curve, decision curve and ROC curve. Conclusion A substantial elevation of serum SPARCL1 levels during the early period after ICH is independently related to illness severity and poor neurological outcomes, thus signifying that serum SPARCL1 may appear as a prognostic biomarker of ICH.
... Azkenik, hevin hainbat tumoretan ere adierazten da (esate baterako, meningiometan, biriketako minbizi ez-zelularrean, kartzinoma gastrikoan eta prostatako eta koloneko kartzinometan) [35,[66][67][68][69]. ...
Article
Full-text available
Proteina matrizelularrak zelulaz kanpoko matrizeko (ZKM) molekulak dira, zeinek beste ZKMko molekuletatik bereizten dituzten funtzio espezifikoak baitituzte. Adibidez, zelulen funtzioa modulatzen dute eta gaitasun desitsaskorrak dituzte, besteak beste. Azkenengo urteetan, hainbat molekulek proteina matrizelularren ezaugarriak betetzen dituztela ikusi da, eta horien parte-hartzea nabarmenduz joan da gaixotasun neuropsikiatrikoetan. Lan honetan, alde batetik, proteina matrizelularren ezaugarriak eta talde honetako partaide nagusien funtzio garrantzitsuenak azalduko dira. Eta, bestetik, gehiago sakonduko da hevin proteina matrizelularraren funtzioetan, gaixotasun neuropsikiatrikoetan duen inplikazioa aipatuz.
... To further elucidate whether hevin mRNA increase paralleled an enhancement in the translation process, the protein expression levels were also determined. Both previously described~130 kDa and~100 kDa hevin bands [22,31,41,42] were measured in total homogenates of PFC, HIP, CAU and CB. In accordance with the alterations observed in hevin mRNA, an increase in the~130 kDa and~100 kDa forms was detected in PFC and CB of AUD subjects in comparison to controls (PFC: 25% increase in~130 kDa band, p = 0.0015 and 46% increase in~100 kDa band, p < 0.0001; CB: 32% increase in~130 kDa band, p = 0.0344 and 38% increase in~100 kDa band, p = 0.013) (Figure 2A,D). ...
Article
Full-text available
Astrocytic-secreted matricellular proteins have been shown to influence various aspects of synaptic function. More recently, they have been found altered in animal models of psychiatric disorders such as drug addiction. Hevin (also known as Sparc-like 1) is a matricellular protein highly expressed in the adult brain that has been implicated in resilience to stress, suggesting a role in motivated behaviors. To address the possible role of hevin in drug addiction, we quantified its expression in human postmortem brains and in animal models of alcohol abuse. Hevin mRNA and protein expression were analyzed in the postmortem human brain of subjects with an antemortem diagnosis of alcohol use disorder (AUD, n = 25) and controls (n = 25). All the studied brain regions (prefrontal cortex, hippocampus, caudate nucleus and cerebellum) in AUD subjects showed an increase in hevin levels either at mRNA or/and protein levels. To test if this alteration was the result of alcohol exposure or indicative of a susceptibility factor to alcohol consumption, mice were exposed to different regimens of intraperitoneal alcohol administration. Hevin protein expression was increased in the nucleus accumbens after withdrawal followed by a ethanol challenge. The role of hevin in AUD was determined using an RNA interference strategy to downregulate hevin expression in nucleus accumbens astrocytes, which led to increased ethanol consumption. Additionally, ethanol challenge after withdrawal increased hevin levels in blood plasma. Altogether, these results support a novel role for hevin in the neurobiology of AUD.
... SPARC's fulllength protein appears with a relative mass of approximately 75 kDa and 150 kDa in vitro, suggesting that it might be expressed in the form of monomer or homodimer. SPARCL1 is a protein from a single-copy gene and has three major highly conserved domains, which are an N-terminal acidic calcium binding domain, a follistatin-like structure with a central part rich in cysteines, and a C-terminal extracellular Ca 2+ -binding domain [63][64][65][66]. A process of proteolysis by specifically matrix metalloproteinase-3 (MMP-3) and ADAMTS4, which are a disintegrin and metalloproteinase with thrombospondin motifs 4, is involved in the activation of SPARCL1 protein [67,68]. ...
Thesis
Full-text available
Objectives: Colorectal carcinoma (CRC) is the second most common cancer for women and the third most common cancer for men in Germany. The primary cause of death in CRC patients is metastatic disease and its corresponding complications. At the molecular level, specific tumor microenvironments (TMEs) and the genotypic diversity of tumor cells influence the plasticity and heterogeneity of both tumor and stromal cells, and in consequence tumor progression and malignancy. Previous results showed that in tumor endothelial cells (TECs), which were isolated from CRC tissues with a TME associated with improved clinical prognosis, the expression of SPARCL1 was upregulated and of EDIL3 and HEY2 downregulated compared to TECs isolated from CRC with worse clinical prognosis. Whether this TME-associated differential gene expression of TEC can be confirmed at the tissue level remained unclear. Materials and methods: In this work the expression of the above mentioned genes has been investigated in whole tumor extracts from primary CRC lesions, hepatic metastases and corresponding normal tissues using RT-PCR. Observations and results: The expression levels of SPARCL1, EDIL3 and HEY2 in whole extracts of primary colorectal carcinoma were downregulated compared to corresponding normal colon tissue at the RNA level (n=29, p < 0.001 for SPARCL1 and EDIL3; p < 0.01 for HEY2). In corresponding to liver tissue, the expression of SPARCL1 (p < 0.01) and HEY2 (p < 0.001) was lower and of EDIL3 higher (p < 0.001) in metastatic CRC lesions compared to the corresponding normal liver tissue. Conclusions: Our results show that the downregulation of SPARCL1 expression between isolated normal endothelial cells (NECs) and TECs is still present when comparing whole normal colon and CRC tissues, respectively. In fact, SPARCL1 was mainly expressed in endothelial cells. Moreover, a loss of SPARCL1 expression was also detected in metastatic CRC liver lesions. In contrast, the overall expression levels of EDIL3 and HEY2 were not in accordance with their expression in endothelial cells, but likely altered by other EDIL3- and HEY2-expressing cells in the whole cell extracts. Different independent techniques such as RNAscope, IHC or Western Blot are required to further decipher the overall and cell-specific expression levels of SPARCL1, EDIL and HEY2 in colorectal carcinoma and thereby get insights in their role in this disease.
... SPARC has been reported to play important roles in skeletal muscle myoblast differentiation [4,5]. SPARCL1 (SPARC-like protein 1, also known as ECM2, Hevin, MAST 9, or SC1), also belongs to SPARC family of matricellular proteins, it shares a follistatin-like module and an extracellular Ca 2+ binding domain in that of SPARC in mouse [6]. A report showed that SPARCL1 and SPARCL1 may share the similar biological function such as the antagonist of cell adhesion [3]. ...
Article
Full-text available
As an extracellular matrix protein, secreted protein acidic and rich in cysteine (SPARC)-like 1 (SPARCL1) is involved in various cell functions. It was previously implicated in bovine skeletal muscle-derived satellite cell (MDSC) differentiation; however, the underlying mechanism remains unknown. In this study, immunoprecipitation and mass spectrometry revealed that integrin β1 (ITGB1) combines with SPARCL1. Further, co-immunoprecipitation demonstrated that SPARCL1 interacts with ITGB1. Cell scratch assays explored the influence of SPARCL1 on MDSC migration through ITGB1. In addition, desmin staining for myotube fusion rate and MyoD protein expression results showed that SPARCL1 promotes MDSC early differentiation through ITGB1. Furthermore, Western blotting results demonstrated that SPARCL1 regulates the expression of p-FAK, p-paxillin, vinculin, Cdc42, and Arp2/3 through ITGB1. These findings indicate that SPARCL1 may influence bovine MDSC migration and differentiation through an ITGB1-mediated cell signaling pathway. Herein, we elucidated the mechanism through which SPARCL1 affects MDSC differentiation. Our results provide insight into the molecular mechanism of muscle development and may in the future facilitate skeletal muscle regeneration and treatment.
... SPARCL1 was originally screened out from a brain cDNA expression library by Johnston et al. 2 . The SPARCL1 protein, which is approximately 650 amino acids, can be divided into three major components: N-terminal acidic domain, Follistatinlike domain (FS), and C-terminal extracellular calciumbinding domain 3 . SPARC has been demonstrated to regulate biological processes such as cell proliferation 4 , anticell adhesion 5 , and tissue repair 6 . ...
Article
Full-text available
The extracellular matrix (ECM) is known to regulate tissue development and cell morphology, movement, and differentiation. SPARCL1 is an ECM protein, but its role in mouse cell differentiation has not been widely investigated. The results of western blotting and immunofluorescence showed that SPARCL1 is associated with the repair of muscle damage in mice and that SPARCL1 binds to bone morphogenetic protein 7 (BMP7) by regulating BMP/transforming growth factor (TGF)-β cell signaling. This pathway promotes the differentiation of C2C12 cells. Using CRISPR/Cas9 technology, we also showed that SPARCL1 activates BMP/TGF-β to promote the differentiation of C2C12 cells. BMP7 molecules were found to interact with SPARCL1 by immunoprecipitation analysis. Western blotting and immunofluorescence were performed to verify the effect of BMP7 on C2C12 cell differentiation. Furthermore, SPARCL1 was shown to influence the expression of BMP7 and activity of the BMP/TGF-β signaling pathway. Finally, SPARCL1 activation was accompanied by BMP7 inhibition in C2C12 cells, which confirmed that SPARCL1 affects BMP7 expression and can promote C2C12 cell differentiation through the BMP/TGF-β pathway. The ECM is essential for muscle regeneration and damage repair. This study intends to improve the understanding of the molecular mechanisms of muscle development and provide new treatment ideas for muscle injury diseases.
... Hevin is structurally the closest relative of SPARC and the two proteins have many overlapping roles. Like SPARC, Hevin can be anti-adhesive, modulates cell shape, binds to collagen and regulates collagen assembly [42][43][44][45] . While the role of Hevin in diabetes has not yet been examined, Hevin has been implicated in many cancers. ...
... High molecular weight Hevin-reactive bands have also been previously observed [44][45][46]86 . Based on the number of predicted glycosylation sites (Suppl. ...
... Figure 1). It has been suggested that this SPARC-like fragment may compensate for the loss of SPARC expression 44,46,89 . SPARC can be suppressed for example as a result of SPARC promoter methylation during tumourigenesis 90 . ...
Article
Full-text available
SPARC is a matricellular protein that is involved in both pancreatic cancer and diabetes. It belongs to a wider family of proteins that share structural and functional similarities. Relatively little is known about this extended family, but evidence of regulatory interactions suggests the importance of a holistic approach to their study. We show that Hevin, SPOCKs, and SMOCs are strongly expressed within islets, ducts, and blood vessels, suggesting important roles for these proteins in the normal pancreas, while FSTL-1 expression is localised to the stromal compartment reminiscent of SPARC. In direct contrast to SPARC, however, FSTL-1 expression is reduced in pancreatic cancer. Consistent with this, FSTL-1 inhibited pancreatic cancer cell proliferation. The complexity of SPARC family proteins is further revealed by the detection of multiple cell-type specific isoforms that arise due to a combination of post-translational modification and alternative splicing. Identification of splice variants lacking a signal peptide suggests the existence of novel intracellular isoforms. This study underlines the importance of addressing the complexity of the SPARC family and provides a new framework to explain their controversial and contradictory effects. We also demonstrate for the first time that FSTL-1 suppresses pancreatic cancer cell growth.
... By the time of disease onset, P90, astroglial expression of SPARCL1 was found to have the most significant downregulation in ALS. SPARCL1 (synaptic cleft-1, SC1, MAST-9, RAGS1, ECM2, hevin) is a member of the SPARC (Secreted Protein Acidic and Rich in Cysteine) family of matricellular proteins that modulate cell-extracellular matrix interactions (Brekken, 2004;Johnston, Paladino, Gurd, & Brown, 1990). Members of this protein family contain an acidic domain l, a follistatin-like domain, and an extracellular calcium-binding (E-C) domain containing a pair of Ca 2þ binding EF-hand motifs (Yan & Sage, 1999). ...
Article
Astroglia are a morphologically diverse and highly abundant cell type in the CNS. Despite these obvious observations, astroglia still remain largely uncharacterized at the cellular and molecular level. In disease contexts such as amyotrophic lateral sclerosis (ALS), it has been widely shown that astroglia downregu- late crucial physiological functions, become hypertrophied, reactive, and toxic to motor neurons. However, little is known about the astroglia-specific transcriptomic changes that occur during ALS dis- ease progression, especially early in disease. To address this, we FACS-isolated pure astroglia from early and mid-symptomatic superoxide dismutase 1 (SOD1) G93A spinal cord and performed microarray sequencing, in hopes to uncover markers and pathways driving astroglia dysfunction in ALS. After extensive analyses, we uncovered genes selectively enriched and downregulated in both control and SOD1 astroglia at both disease points. In addition, we were able to identify genes and pathways differ- entially expressed that may have relevance with other neurodegenerative diseases, such as Parkinson’s and Alzheimer’s disease, suggesting a common theme among astroglial dysfunction in neurodegenera- tive disease. In aggregate, this study sheds light on the common and unique themes of dysfunction that astroglia undergo during neurodegenerative disease progression and provides candidate targets for therapeutic approaches.
... In regards to lung and pancreatic tumor xenografts, Brekken and colleagues found, on immunohistochemical analysis, a specific and selective reactivity of monoclonal antibodies directed against SPARCL1 not only in stromal and endothelial cells, but also in tumor cells (Brekken and Sage, 2001;Brekken et al., 2004). ...
... Beside the de-adhesive activity of SPARCL1, it was histologically found to be localized in tumor area involved in desmoplastic transformation (Brekken and Sage, 2001;Brekken et al., 2004). ...
Article
Matricellular proteins are secreted, nonstructural proteins, involved in the mediation of molecular interactions between cells and extracellular microenvironment. They include several, structurally unrelated, members and their homologs. Among these a particularly interesting one is SPARCL1 due to its potential interactions in tumor biology. SPARCL1 is a secreted glycoprotein, belonging to SPARC family of matricellular proteins. It is implicated in the regulation of cell adhesion, migration, and proliferation. SPARCL1 is expressed in physiological context, both during embryogenesis and in adult life during tissue remodeling. Its diverse expression pattern in different forms of human cancers has suggested it may play different roles in tumor biology, as both oncogene and tumor suppressor, based on tumor type. Aim of this review is to critically revise current knowledges about the role, played by SPARCL1, in physiological and pathological contexts and highlight its role as a key-gene in the regulation of tumor biology.
... While SPARCL1 has been shown to inhibit in vitro proliferation of colon cancer (6) and HeLa (8) cells, other studies support that SPARCL1 may not regulate cellular proliferation in the prostate (4,7). Alternatively, SPARCL1 has been shown in multiple models to inhibit processes integral to both local and metastatic progression, such as cancer cell adhesion, migration, and invasion (4,6,7,9,10). Two recent reports demonstrate that SPARCL1 suppresses tumor nodule formation in visceral organs following intravenous injection (6,7). ...
Article
Full-text available
Prostate cancer is a leading cause of cancer death in men due to the subset of cancers that progress to metastasis. Prostate cancers are thought to be hardwired to androgen receptor (AR) signaling, but AR-regulated changes in the prostate that facilitate metastasis remain poorly understood. We previously noted a marked reduction in Secreted protein, acidic and rich in cysteine-like 1 (SPARCL1) expression during invasive phases of androgen-induced prostate growth, suggesting that this may be a novel invasive program governed by AR. Herein, we show that SPARCL1 loss occurs concurrently with AR amplification or overexpression in patient based data. Mechanistically, we demonstrate that SPARCL1 expression is directly suppressed by androgen-induced AR activation and binding at the SPARCL1 locus via an epigenetic mechanism, and these events can be pharmacologically attenuated with either AR antagonists or HDAC inhibitors. We establish using the Hi-Myc model of prostate cancer that in Hi-Myc/Sparcl1-/- mice, SPARCL1 functions to suppress cancer formation. Moreover, metastatic progression of Myc-CaP orthotopic allografts is restricted by SPARCL1 in the tumor microenvironment. Specifically, we show that SPARCL1 both tethers to collagen in the extracellular matrix (ECM) and binds to the cell's cytoskeleton. SPARCL1 directly inhibits the assembly of focal adhesions thereby constraining the transmission of cell traction forces. Our findings establish a new insight into AR-regulated prostate epithelial movement and provide a novel framework whereby, SPARCL1 in the ECM microenvironment restricts tumor progression by regulating the initiation of the network of physical forces that may be required for metastatic-invasion of prostate cancer. Copyright © 2015, American Association for Cancer Research.