Skip to main content
Erschienen in: Journal of Mammary Gland Biology and Neoplasia 2/2013

Open Access 01.06.2013

Positional Variations in Mammary Gland Development and Cancer

verfasst von: Jacqueline M. Veltmaat, Ann F. Ramsdell, Esta Sterneck

Erschienen in: Journal of Mammary Gland Biology and Neoplasia | Ausgabe 2/2013

Abstract

Most mammals develop their mammary glands in pairs of which the two counterparts are symmetrically displaced away from the ventral midline. Based on this symmetry and the same functional outcome as a milk-producing organ, the mammary glands are easily presumed to be mere copies of one another. Based on our analysis of published data with inclusion of new results related to mammary development and pathology in mice, we argue that this presumption is incorrect: Between and within pairs, mammary glands differ from one another, and tumor incidence and biology depend on the position along the anterior-posterior and the left-right axis as well. This insight has implications for experimental designs with mouse models and for data extrapolation between mammary glands within and between species. We suggest that improved documentation of location-specific mammary gland features will lead to more insights into the molecular mechanisms of mammary gland development and cancer biology in both mice and humans.
Abkürzungen
AP
anterior-posterior
DMBA
7,12-demethylbenz-alpha-anthracene
DV
dorso-ventral
LR
left-right
LPM
lateral plate mesoderm
MFP
mammary fat pad
MG
mammary gland
MR
mammary rudiment
RA
retinoic acid
TEB
terminal end bud

Introduction

Within the animal Class of Mammals the number of mammary glands (MGs) varies considerably, from two in most primates to 25 in some opossums [1]. Mammary glands emerge along bilateral mammary lines, that are in some species distinct as ridges in the surface ectoderm, one on each flank of the embryo [2]. In mouse embryos, the mammary lines do not form ridges, yet become histologically and molecularly distinct between embryonic day (E) 10.5 and E11.5 (Fig. 1), and yield five mammary rudiments (MRs) on each side of the embryo by E12 [3]. A few species have a double row of mammary glands on each body half [1].
Studies in the rabbit led to the contention that mammary line formation is completed prior to mammary placode formation, per flank providing a shared ‘anatomical’ origin for all mammary glands arising on that flank. Moreover, other repetitive structures along the anterior-posterior (AP) body axis, such as limbs, are induced by the same molecular mechanism. Based on those two contentions, one might presume that all MRs are induced via shared molecular mechanisms.
However, we now know that MRs arise prior to completion of mammary line formation in the mouse, and not in sequence along the AP-axis [3], which suggests that the presumption that the mammary line is a pre-existing structure from which the mammary glands subsequently derive by a shared mechanism, is incorrect, at least for mice [4]. Moreover, defects in the formation and morphogenesis of select mammary gland pairs in various genetically modified mice show that different signaling networks induce and maintain mammary gland development at different positions along the AP-axis. Furthermore, the reciprocal tissue interactions governing mammary gland development [5] may co-vary with the high internal left-right (LR) asymmetry of the trunk. Indeed, LR-asymmetry between counterparts of mammary gland pairs has also become evident.
Thus, each mammary gland has an individual identity. Future studies on normal and pathological mammary gland development will benefit greatly from consistent documentation and analysis of data relative to mammary gland position. Accordingly, we will refer to them by number 1–10, as opposed to pair (1–5) or position (Fig. 1), unless the cited literature refers to mammary glands as clusters (e.g. anterior, posterior) without explicit specification of which glands were subject to examination.

Molecular Variation in the Induction of a Mammary Cell Fate Along the Mammary Lines

What determines the number and position of the individual pairs of mammary glands? The 2:1 correlation between the number of mammary glands and average litter size in most species [6] and co-incidence of supernumerary nipples with more frequent twinning as reported for isolated groups of sheep and macaques [7, 8] suggests considerable genetic plasticity that is molded by evolutionary pressure. Indeed, supernumerary breasts (polymastia) or nipples (polythelia) are an inherited trait in some humans [9], and pigs and sheep can be selectively bred for increased teat number [7, 10], indicating the involvement of a genetic component. Accordingly, several genetically modified mice have been identified with mammary induction defects leading to numerical aberrancies (Table 1). They can be organized into four categories depending on the subset of MRs affected, and loss or gain of MRs.
Table 1
Position-dependent mammary induction defects as established at E11.5 or E12.5
https://static-content.springer.com/image/art%3A10.1007%2Fs10911-013-9287-3/MediaObjects/10911_2013_9287_Tab1_HTML.gif
Legend: https://static-content.springer.com/image/art%3A10.1007%2Fs10911-013-9287-3/MediaObjects/10911_2013_9287_Figa_HTML.gif MR absent; https://static-content.springer.com/image/art%3A10.1007%2Fs10911-013-9287-3/MediaObjects/10911_2013_9287_Figb_HTML.gif MR present without reported anomaly; https://static-content.springer.com/image/art%3A10.1007%2Fs10911-013-9287-3/MediaObjects/10911_2013_9287_Figc_HTML.gif MR hypoplastic; https://static-content.springer.com/image/art%3A10.1007%2Fs10911-013-9287-3/MediaObjects/10911_2013_9287_Figd_HTML.gif MR severely hypoplastic; https://static-content.springer.com/image/art%3A10.1007%2Fs10911-013-9287-3/MediaObjects/10911_2013_9287_Fige_HTML.gif MR forms late and hypoplastic; https://static-content.springer.com/image/art%3A10.1007%2Fs10911-013-9287-3/MediaObjects/10911_2013_9287_Figf_HTML.gif small supernumerary MR close to normal MR; https://static-content.springer.com/image/art%3A10.1007%2Fs10911-013-9287-3/MediaObjects/10911_2013_9287_Figg_HTML.gif large MR. The region between MR3/8 and MR4/9 is prone to formation of supernumerary MRs of variable size (category IV)
hypo hypomorphic mutant; n.d not determined; v.p. variable penetrance for presence, size and timing of formation
I
Loss of subset, linear sequence. The only example in this category is the transcription factor Hoxc6. Its loss occasionally leads to smaller thoracic buds at E12.5, and consistently to reduced fat pad development and branching morphogenesis by birth. These defects correspond to the Hoxc6 expression domain in the anterior part of the body, and are consistent with the general functions of Hox genes in providing and interpreting AP-positional information.
 
II
Loss of subset, non-linear sequence. This cluster contains a variety of transcription (Gli3, Pax3, Tbx2, Tbx3) and secreted factors (Fgf10, Nrg3). Interestingly, their expression in the somites or somite-derived dermal mesenchyme, and in particular somitic Fgf10 expression finally gave an identity to so-called ‘dermal factors’, which had long before been postulated to confer the mammary gland-inducing potential of the dermis [11, 12]. The dissimilarities in phenotypes of the mouse mutants in this category indicate that different molecular mechanisms for mammary induction exist along the AP-axis, and that the five mammary gland pairs develop independently of each other.
 
III
Loss of all MRs (with variable penetrance). Most genes in this category (Dkk1, Lef1, Pygo2, Lrp4-6, Wise) participate in the initial ectodermal Wnt signaling. The phenotypic clustering of their mutations suggests that the various mesodermal mammary inducers all converge on the same signaling cascade(s) in the ectoderm.
 
IV
Supernumerary MR formation. Increased ectodermal Ectodysplasin-A1 or Wnt signaling leads to formation of supernumerary glands along the mammary line, showing that the mammogenic capacity is maintained along the entire mammary line. Moreover, increased Wnt signaling leads to larger MRs, and in the case of reduced Wise/Sostdc1 function, a reduced spacing between MR2/7 and MR3/8 as well. The latter can be attributed to more ectodermal cells adopting a mammary fate and consequently losing proliferative activity that is required to fill the space between these MR pairs. A supernumerary MR slightly dorsal to the normal MR4 or MR9 is transiently present in some wild-type embryos of mixed genetic backgrounds but persists in some Nrg3 Ska mutants, showing that mammogenic potential also exists at atypical positions outside the line.
 
The various mesenchymal inductive mechanisms seem to converge on shared epithelial regulators, whose extent of activation appears to regulate the number of induced MRs. It is conceivable then that the variable inductive signals bestow the MR cells with unique properties. Indeed, preliminary gene profiling data suggest that each of the five paired mammary placode epithelia express unique subsets of genes (Sun and Veltmaat, http://​www.​veltmaatlab.​net/​research.​html). While the functional relevance of these genes remains to be determined, mammary development proceeds differently in all MRs, as evidenced by the different growth rate and morphogenesis of the MRs [13], culminating in rudimentary glandular trees before birth that differ in size and number of branches (Fig. 2). These differences may very well underlie some of the differential attributes of adult MGs (discussed below).
Why do mesodermal genes act differently at different positions along the mammary line? The answer may be found in their expression pattern, which is in part reflected in the temporal development of the murine mammary line. This line is actually composed of three initially unconnected streaks of Wnt10b expression [3]. As concluded from the phenotypes summarized in Table 1, these streaks form independently of each other.
The first streak forms on the flank, between the limbs. Its initially fragmented Wnt10b expression pattern in wild-type embryos (Fig. 1) and dorsalized position upon hypaxial truncation of the somites, as in Pax3 null mutants, indicate somitic involvement in the formation of this streak [14]. Fgf10 is expressed highly in the hypaxial dermomyotomal tips of somites #12–18, which underlie the region where MR2/7 and MR3/8 form [14, 15]. FGF10 activates its main receptor FGFR2-IIIb, expressed throughout the overlying surface ectoderm. Partial reduction of somitic Fgf10 expression, as in Gli3 Xt-J/Xt-J null or hypomorphic Fgf10 mlcv/- mutants, allows formation of a mammary line and all MRs except MR3/8, indicating that at least this pair requires somitic Fgf10 and, contrary to MR2/7, is susceptible to reduced levels of Fgf10 [14]. Fgf10 is not expressed in somites 19–24, and neither Wnt10b expression in this region nor induction of MR4/9 (above #24 somites) requires FGF10/FGFR2b signaling. These observations suggest that the anterior and posterior regions of this streak are two separate entities which develop by independent mechanisms, and that another yet to be identified somitic molecule(s) may be responsible for the initiation of mammogenesis in the posterior region, at the posterior edge of which arises MR4 or MR9. Interestingly, the boundary between the Fgf10-dependent and Fgf10-independent region at the level of #18 somites is home to a small mesenchymal Nrg3 expression domain, which seems a prerequisite for normal formation of MR3/8 [16].
The two other mammary streaks are located in the axilla and inguen, where they give rise to MR1/6 and MR5/10, respectively. They gradually elongate dorsally until they connect to the first streak on the flank [3]. Defects in these streaks or their MRs often coincide with defects in limb induction or specification in the absence of limb mesenchymal factors like Tbx2, Tbx3, Gli3 and Fgf10 [13, 14, 17, 18]. It remains to be determined whether MR1/6 and MR5/10 require such factors or the limb mesenchyme directly, or indirectly via formation of the limbs as new signaling centers. Perhaps MR2/7, that form at the junction of the first and axillary streak, and depend less than MR3/8 on somitic signals [14], similarly benefit from signals that originate in the limb. In conclusion, MRs develop due to in part variable mesodermal inductive signals along the AP-axis that converge on common ectodermal executive pathways. Thus, the different glands experience unique developmental histories and, furthermore, are embedded and surrounded by tissue that exhibits region-specific properties.

Left-Right Asymmetry During Embryonic Mammary Development

The embryonic flanks develop in the trunk as two broad lateral regions where the dorsal paraxial mesoderm intermingles with the ventral lateral plate mesoderm (LPM). They establish new, ventro-lateral boundaries with the ventrum, which serve as bilateral platforms for mammary line formation. Externally, these presumptive mammary forming regions look left-right (LR) symmetric. But looks can be deceiving! LR asymmetries during mammogenesis become evident at the level of individual mammary gland pairs: The size and branching pattern differ between contralateral glands; one counterpart of a normal or supernumerary pair may be missing or shifted from its symmetric position, shown for mouse in Figs. 2 and 3. In humans with Poland syndrome, one breast (most often the right) may remain hypoplastic [19]. Interestingly, formation of the mammary line and MRs is slightly advanced on the left side (Fig. 3) and the left side seems to have a greater mammogenic potential. For example, unilateral polythelia or polymastia occurs most frequently on the left side in humans [20] and mice [21].
The basis for LR asymmetric mammary gland development is likely rooted in embryonic LR patterning: During vertebrate embryogenesis, the LR axis establishes the positions of the heart and visceral organs and promotes structural asymmetries that are necessary for their functions. The driving force for LR patterning is unilateral activity of molecules such as the secreted factor nodal, which is expressed by the left half of the node, left-sided LPM and overlying ectoderm [22]. In the left-sided LPM, nodal initiates the expression of the transcription factor Pitx2, an essential “interpreter” of LR axial patterning during organogenesis [reviewed by 23]. Thus, as ectodermal appendages that are induced at the ventro-lateral boundaries, i.e. at the fronts of somitic penetration into two molecularly different regions of left and right LPM [14], the left and right mammary glands have distinct molecular “histories” from their inception!
Moreover, and despite their morphological bilateral symmetry, the left and right somites also differ from each other with significant LR differences in gene expression [24, 25]. Of interest is perhaps heparin-binding EGF-like growth factor [25], which is transiently elevated on the left side, temporally overlapping with formation of MR3/8. Furthermore, retinoic acid (RA) signaling promotes LR asymmetry in the somites, as demonstrated by one-sided delay in somite formation in chick and zebrafish embryos, caused by pharmacological inhibition of RA production [26, 27]. Several modulators of RA signaling and synthesis, including Raldh2, are expressed in the somites proper, and the RA receptor RARβ is expressed in the flank mesenchyme underlying MR2/7, 3/8, 4/9 [28]. Notably, perturbation of RA-signaling affects MR3/8 formation ex vivo [28]. Unfortunately, laterality in effects of RA signaling on mammary gland development was not tested. This will be difficult to do, due to lethality of RA-deprived mouse models prior to mammary gland formation, and due to artifacts introduced during MR development ex vivo under otherwise normal conditions, but especially under experimental conditions such as pharmological modulation of RA-signaling. However, it is of interest that RA-deficient Raldh2 −/− mouse embryos exhibit transiently delayed right-sided development and segmentation of the paraxial mesoderm into somites #8–15 [29]. With MR2/7 and MR3/8 forming above the #12 and #15 somites respectively, it is thus conceivable that RA signaling may be a pivotal link between somitic laterality and asynchrony and asymmetry in development of LR counterparts of particularly these MR pairs. Moreover, the already known requirement of somitic signals for induction of MR3/8 [14], provides strong support for the hypothesis that this mammary gland pair—despite its seemingly morphological symmetry - may be lateralized, either directly or indirectly via inductive interaction with lateralized aspects of somites, including effects of RA-signaling.
Finally, the curvature of the AP-body axis may also be involved in promoting subtle asymmetry of mammogenesis: Most often the AP-axis is curved to the right (tail over right shoulder), leading to a ‘compressed’ right flank and ‘stretched’ left flank. If molecular gradients are involved in early mammary gland formation, which is the case at least for somitic Fgf10 [14], their shape may be different between left and right flanks, which could possibly explain the slight LR asynchrony in length of the mammary lines and mis-alignment between paired glands that is sometimes observed even in wild-type embryos (Fig. 3).

Left-Right Asymmetry in Breast Cancer

A striking but still unexplained feature of breast cancer is a higher tumor incidence in the left breast. Although modest with a ~1.10 left/right incidence ratio, such laterality was consistently found in several epidemiological studies [3036] regardless of gender and stage of disease. Even in bilateral breast cancer cases, the left breast is more frequently affected first, or with a larger tumor [37].
Survival rates also appear to reflect LR differences: A breast cancer study among 590 Russian patients reported a statistically significant higher survival rate of patients with right-sided, advanced stage disease [38]. In contrast, a study of 163 Norwegian patients associated longer survival with left-sided disease, but did not relate this finding to disease stage [39]. In a third study of 10,702 Israeli women, no laterality was detected for overall patient survival from years 1–15 [40], but trends of a higher 5–10 year survival rate of left-sided later stage disease, and a higher 10–15 year survival rate of right-sided early stage disease, call for statistical analysis of the data within such smaller time periods, which had not been done. The most recent study of 384 Pakistani breast cancer patients indicates that although left-sided tumors are larger and more prevalent, when standardized for tumor size, right-sided tumors are more prone to metastasis and are associated with lower survival rates [36]. Although the results of these studies appear discordant with each other, they certainly attest to the need for well-defined parameters to better assess laterality. Furthermore, these available data contest, at least in part, the assumption that asymmetry in survival may be due to radiation-induced heart damage, which is greater with left-side targeted therapy. Instead, the data suggest that the LR differences may be due to lateralized tumor biology, similar to LR asymmetry in incidence and patient survival observed with cancers of other paired organs, including kidney cancer, which does not entail cardiac complications following side-specific radiation therapy [33, 41].
Why do tumors form with unequal probability and perhaps outcome in left versus right members of organ pairs? A variety of epidemiological correlates have been investigated, including age, race, ethnicity, sex, breast size, stage of disease at diagnosis, histological tumor type, and even left- versus right-handedness. None of these factors emerged as a consistently significant variable for the increased left-sided incidence [3032, 37]. By contrast, laterality in outcome may in part be explained by laterality of axillary lymph node metastasis. A higher mean number of lymph nodes with metastases in breast cancer patients has been reported for right-sided tumors, consistent with more lymph nodes present on the right side [43, 44]. However, when axillary lymph node involvement was analyzed with respect to the total number of affected nodes, the right-sided increase was present only in patients with 1–3 positive nodes [45]. For patients with >3 positive nodes (a characteristic of locally advanced or higher stage disease) left-sided tumors were associated with more affected nodes despite the LR asymmetry in overall node number [45]. Thus, metastasis to lymph nodes seems to be a lateralized disease feature, and may indicate LR differences in tumor biology, e.g. greater metastatic potential of left-sided tumors, or lateralized features of the lymph nodes themselves. In general, attempts to identify lateralized features may be confounded by the profound heterogeneity of breast cancer and could possibly be improved by inclusion of more detailed cancer subtype-specific information such as genomic classifiers. Indeed, an interesting correlate in laterality of breast tumors is the expression of Her-2 and perhaps also hormone receptors, although the sidedness may co-vary with other parameters: Among 166 Kuwaiti patients spanning all disease stages at diagnosis, right-sided tumors are significantly associated with amplified Her-2 expression, without lateralized differences for hormone receptor expression [46], whereas among 384 Pakistani breast cancer patients with bone metastasis, right-sided tumors were markedly less prone to express Her-2, estrogen receptor, or progesterone receptor, either individually or in combination [36].
It is tempting to speculate that the LR differences in tumor incidence and patient survival may be related to expression of genes that govern LR axial patterning during embryonic development, like Nodal and Pitx2 [reviewed by 47]. Increased expression of Nodal and methylation of Pitx2 DNA correlates with poor prognostic outcome in breast cancer patients [4850]. Overexpression of another nodal pathway component, the co-receptor Cripto-1, is also associated with decreased patient survival [reviewed by 49]. Consistent with these findings, nodal pathway signaling promotes an invasive phenotype in breast cancer cells and induces tumor vascularization, which has implications for disease progression and relapse [51]. Whether nodal or nodal pathway members are expressed asymmetrically during mammary gland development remains to be investigated, as does the question of whether such putative LR differences are recapitulated in breast tumors.
While the laterality in breast cancer incidence and outcome seems subtle, evidence for its existence is compelling. We may be unaware of more significant aspects of laterality because situs is often not a factor in molecular and clinical correlations, and experimental data in mouse models typically are generated from a single side, or if from both sides, may be combined for analyses, obscuring potential LR differences. To our knowledge, laterality has not been investigated systematically in any mouse tumor mammary model to date. Better insights into laterality of breast cancer incidence, biology, and outcome may lead to a better understanding of the disease(s) and have prognostic value.

“Breast” Cancer in Mouse: Location, Location, Location!

The mouse is widely used as a breast cancer model. The axial position of the human breast approximates the 5th thoracic somite or its derived rib. Taking into account the differences in somite number between mouse and human, this area corresponds best to the area between MG2/7 and 3/8 in the mouse. However, the inguinal mouse MG4/9 are generally used for molecular and histological analyses. Does it matter? As described above, the five pairs of mammary glands in the mouse are each induced by unique inductive signaling networks. Yet surprisingly little information exists on differences between the five pairs of glands in adult mice, both at the level of normal physiology and with respect to response to tumor-initiating events. However, several studies report significantly differential tumor incidence or biology depending on the position along the AP axis. So the answer is almost certainly: Yes!
Examples for AP variations in tumor-incidence and biology come from endogenous tumor agents, chemical carcinogenesis, various treatments, and transgenic approaches. Integration of mouse mammary tumor viruses (MMTV) in the C3H mouse strain causes mammary tumors most often in thoracic glands. While tumor incidences are equal among the gland pairs after correction for total wet tissue weight [52], the relative ratios of epithelial to stromal/adipose tissue may not correspond to wet weights and could confound the analysis. Indeed, increased incidence of DMBA-induced tumors in the thoracic mammary glands of rats was correlated to asynchronous development of the glands. Differences in the number of terminal end buds (TEBs) and alveolar buds per gland results in variable abundance of target cells for chemical carcinogens depending on treatment age [53]. Likewise, murine MG2/7 and MG3/8 exhibit a greater number and larger TEBs than MG4/9 (see also Fig. 2); and MG2/7 specifically undergoes a reduction in TEBs from 5 to 6 weeks of age [54]. The observed asynchrony in postnatal development can also result in differential responses to hormones [53]. These AP-specific developmental features are a plausible basis for increased tumor incidence in thoracic glands. Interestingly, exposure to power frequency magnetic fields (MF) increases DMBA-induced mammary tumorigenesis in SD1 rats, particularly of MG1/6 and 2/7 in correlation with more significant MF-induced increases in ornithine decarboxylase activity—a possible indicator for stem cell proliferation [55]. However, in F433 Fisher rats, MF exposure increased the incidence of DMBA-induced adenocarcinoma specifically in the 5th of the six pairs of glands in rats, but latency was reduced most in the 3rd pair [56]. Thus, region-specific effects on tumorigenesis may vary even between strains of the same species.
Transgenic MMTV-Polyoma middle T antigen (PyMT) causes the largest tumor burden with shortest latency in MG1/6 of C57BL/6 mice [57]. Tumor latency is more similar among all other glands, with MG4/9 harboring the second largest tumor burden. Introduction of an iNOS (Nos2) null mutation increases tumor latency and reduces tumor burden most in MG4/9 and least in MG1/6 [57]. Overexpressed Erbb2 in MMTV-cNeu mice [58] induces tumors preferentially in MG2/7 + 3/8 (Table 2) and tumor incidence is comparatively low in MG4/9/5/10. Transgenic C3(1)-SV40-T/t-antigen causes tumor formation first in MG1/6 and MG2/7 with the highest total tumor burden in MG2/7 [59]. Differential activity of transgene promoters is one possible explanation for variations in transgene-initiated mammary tumorigenesis. Even then, such mechanism(s) would serve to highlight the significance of biological differences between pairs of glands. What is clear from these data, though, is that these transgenic models generate most tumors in specific anterior mammary glands, raising the question if other transgenic models exhibit similar or other unique gland-specific incidences along the AP-axis.
Table 2
Regional distribution of mammary tumors in MMTV-cNeu transgenic mice
Cohort \ Glands
MG1/6
MG2/7+3/8
MG4/9+5/10
A
4 (15)
19 (73)**
3 (12)
B
11 (27)
26 (63)**
4 (10)
Weight (%)
(18)
(46)
(36)
(A) Number of palpable tumors observed in 21 tumor-bearing transgenic mice at the age of 9 months (% of 26 tumors total), and (B) number of tumors as assessed by necropsy in 22 mice at tumor endpoint (% of 41 tumors total). The relative wet weight of the specified glands in % is quoted from [52] and was not specifically determined for this strain. **P < 0.0001 compared to MG4/9 + 5/10 and to MG1/6 by mixed-effects Poisson regression analysis
Transplantation experiments also provide clues to the existence of AP-axial effects both in the tumor cells and environment. PyMT tumor cells isolated from anterior tumors proliferate faster than cells from posterior tumors, independent of implantation site. Significant differences in gene expression exist between anterior and posterior tumors despite equivalent histology, and the difference in proliferative potential was maintained over transplantation passages [60]. Moreover, the higher take rate of teratocarcinoma cell grafts at lumbar subcutaneous sites despite their faster growth rate at thoracic sites [61] provides evidence for unique positional features. Likewise, MDA-MB-435 tumor cell xenografts in thoracic MFPs grow better in response to high fat diet but not when in inguinal MFPs [62]. Thus, along the AP-axis, both the mammary epithelium and stroma harbor positional variations that influence tumor growth.
Finally, modality of disease progression may also depend on positional differences: Tumors of C3HxVY-Avy mice are more likely to metastasize from thoracic glands than from abdominal glands [63]. MDA-MB-435 cell implants into MFPs of athymic nude mice showed the same tendency and moreover, thoracic implants spread via the vascular system, while inguinal implants spread via the lymphatic system [62].
What could be the cause of these positional variations? A comprehensive review article on wound healing, immune responsiveness, and mammary tumorigenesis in mice and rats, concluded that not only tumor growth, but also wound healing responses are more pronounced in anterior regions [64]. The prominent role of inflammatory signaling in breast cancer suggests that this AP-gradient may influence murine mammary tumor biology. The AP-gradient of the nervous system and vascularization, resulting in reduced blood flow in the posterior body, provides perhaps the likeliest explanation for the region-specific variations in tumor biology [64]. Such a gradient has also been confirmed for blood flow in pectoral versus inguinal mammary glands [65]. Consequently, one can hypothesize that posterior glands exhibit reduced oxygen tension and lower concentrations of systemic factors, which may modulate tumor incidence. Alternatively, varying relative positions of lymph nodes may alter lymph drainage of the tissue and consequently tumor biology.
The role of embryonic signaling pathways in promoting cancer cell stemness and metastasis [66] may suggest that differential intrinsic features of tumor cells and their microenvironment could possibly be traced back to the differential developmental signals along the mammary line of the mouse. For example, by interpretation of regional cues, Hox transcription factors lay down positional memory that results in region-of-origin specific cell biology, as shown for dermal fibroblasts [67]. By analogy, mammary stroma may secrete unique factors depending on their position along the AP axis. Aberrant Hox gene activity has also been implicated in breast cancer [68] and in altered Wnt signaling [69, 70]. Given the prominent role of the Wnt pathway in embryonic MG development and in breast tumor biology, it is conceivable that the Hox and Wnt pathways affect tumors differentially along the AP axis.

Conclusions and Perspectives

The mouse is an invaluable model system to study mammalian development, physiology and cancer. Here, we reviewed evidence that each of the ten mammary glands in mouse is unique, from inception to maintenance, throughout normal and tumor development as well as metastasis. Mechanistic insights into causes for variations in prenatal development of the mammary glands begin to emerge. One can presently only speculate about the cause(s) for regional variations in tumor incidence and biology, which may in part stem from intrinsic molecular differences during embryogenesis, and from variations in stromal factors including vascularization and lymph drainage. However, this phenomenon can possibly be exploited to gain insights into factors that modulate tumorigenesis. Clearly, future studies on normal and pathological mammary gland development will benefit greatly from improved documentation of positional variations. In particular, laterality should be treated as a possible variable in studies of human patients to discern why tumor incidence and progression are lateralized disease features and to assess the potential implications for the clinic. Inbred mouse strains and engineered mouse models provide unique opportunities to study positional variations from both left-right and anterior-posterior axis effects. Invaluable information may be lost when samples/data from different locations are combined before analyses, or confounded when phenotypic aspects of one gland are correlated to molecular data from another. Moreover, experimental design and analysis of specific cancer models may improve when regional effects are taken into account, e.g. by tailoring treatments to developmental asynchrony and by making guided choices such as using inguinal glands for studies of lymphatic metastasis. Ultimately, such consideration of positional variations in mammary gland development and tumorigenesis should improve the predictive value of studies in mouse for various aspects of human breast cancer.

Acknowledgments

We are indebted to many colleagues for suggestions and discussion. We thank Glenn Summers, Robert Leighty, Thu Duong, May Yin Lee and Li Sun for expert assistance. Research was supported by A*STAR (J.M.V.), National Institute of Health (NIH) HD068993 (A.F.R.) and the Intramural Research Program of the NIH in part with Federal Funds under contract no.HHSN261200800001E (E.S).
Open Access This article is distributed under the terms of the Creative Commons Attribution 2.0 International License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Gynäkologie

Kombi-Abonnement

Mit e.Med Gynäkologie erhalten Sie Zugang zu CME-Fortbildungen der beiden Fachgebiete, den Premium-Inhalten der Fachzeitschriften, inklusive einer gedruckten gynäkologischen oder urologischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Bresslau E. The structure and development of the mammary apparatus in the marsupialia., in The Mammary Apparatus of the Mammalia—in the light of ontogenesis and phylogenesis. London: Methuen and Co. Ltd; 1920. p. 44–95.CrossRef Bresslau E. The structure and development of the mammary apparatus in the marsupialia., in The Mammary Apparatus of the Mammalia—in the light of ontogenesis and phylogenesis. London: Methuen and Co. Ltd; 1920. p. 44–95.CrossRef
2.
Zurück zum Zitat Propper AY. Wandering epithelial cells in the rabbit embryo milk line. A preliminary scanning electron microscope study. Dev Biol. 1978;67(1):225–31.PubMedCrossRef Propper AY. Wandering epithelial cells in the rabbit embryo milk line. A preliminary scanning electron microscope study. Dev Biol. 1978;67(1):225–31.PubMedCrossRef
3.
Zurück zum Zitat Veltmaat JM et al. Identification of the mammary line in mouse by Wnt10b expression. Dev Dyn. 2004;229(2):349–56.PubMedCrossRef Veltmaat JM et al. Identification of the mammary line in mouse by Wnt10b expression. Dev Dyn. 2004;229(2):349–56.PubMedCrossRef
4.
5.
Zurück zum Zitat Robinson GW, Karpf AB, Kratochwil K. Regulation of mammary gland development by tissue interaction. J Mammary Gland Biol Neoplasia. 1999;4(1):9–19.PubMedCrossRef Robinson GW, Karpf AB, Kratochwil K. Regulation of mammary gland development by tissue interaction. J Mammary Gland Biol Neoplasia. 1999;4(1):9–19.PubMedCrossRef
6.
Zurück zum Zitat Gilbert AN. Mammary number and litter size in Rodentia: the “one-half rule”. Proc Natl Acad Sci U S A. 1986;83(13):4828–30.PubMedCrossRef Gilbert AN. Mammary number and litter size in Rodentia: the “one-half rule”. Proc Natl Acad Sci U S A. 1986;83(13):4828–30.PubMedCrossRef
7.
Zurück zum Zitat Bell A. Bell on the development by selection of supernumerary mammae in sheep. Science. 1899;9(227):637–9.CrossRef Bell A. Bell on the development by selection of supernumerary mammae in sheep. Science. 1899;9(227):637–9.CrossRef
8.
Zurück zum Zitat Hsu MJ et al. High incidence of supernumerary nipples and twins in formosan macaques (Macaca cyclopis) at Mt. Longevity, Taiwan. Am J Primatol. 2000;52(4):199–205.PubMedCrossRef Hsu MJ et al. High incidence of supernumerary nipples and twins in formosan macaques (Macaca cyclopis) at Mt. Longevity, Taiwan. Am J Primatol. 2000;52(4):199–205.PubMedCrossRef
9.
Zurück zum Zitat Cellini A, Offidani A. Familial supernumerary nipples and breasts. Dermatology. 1992;185(1):56–8.PubMedCrossRef Cellini A, Offidani A. Familial supernumerary nipples and breasts. Dermatology. 1992;185(1):56–8.PubMedCrossRef
10.
Zurück zum Zitat Hirooka H et al. A whole-genome scan for quantitative trait loci affecting teat number in pigs. J Anim Sci. 2001;79(9):2320–6.PubMed Hirooka H et al. A whole-genome scan for quantitative trait loci affecting teat number in pigs. J Anim Sci. 2001;79(9):2320–6.PubMed
11.
Zurück zum Zitat Kratochwil K. Organ specificity in mesenchymal induction demonstrated in the embryonic development of the mammary gland of the mouse. Dev Biol. 1969;20(1):46–71.PubMedCrossRef Kratochwil K. Organ specificity in mesenchymal induction demonstrated in the embryonic development of the mammary gland of the mouse. Dev Biol. 1969;20(1):46–71.PubMedCrossRef
12.
Zurück zum Zitat Propper A, Gomot L. Tissue interactions during organogenesis of the mammary gland in the rabbit embryo. C R Acad Sci Hebd Seances Acad Sci D. 1967;264(22):2573–5.PubMed Propper A, Gomot L. Tissue interactions during organogenesis of the mammary gland in the rabbit embryo. C R Acad Sci Hebd Seances Acad Sci D. 1967;264(22):2573–5.PubMed
13.
Zurück zum Zitat Lee MY et al. Ectodermal influx and cell hypertrophy provide early growth for all murine mammary rudiments, and are differentially regulated among them by Gli3. PLoS One. 2011;6(10):e26242.PubMedCrossRef Lee MY et al. Ectodermal influx and cell hypertrophy provide early growth for all murine mammary rudiments, and are differentially regulated among them by Gli3. PLoS One. 2011;6(10):e26242.PubMedCrossRef
14.
Zurück zum Zitat Veltmaat JM et al. Gli3-mediated somitic Fgf10 expression gradients are required for the induction and patterning of mammary epithelium along the embryonic axes. Development. 2006;133(12):2325–35.PubMedCrossRef Veltmaat JM et al. Gli3-mediated somitic Fgf10 expression gradients are required for the induction and patterning of mammary epithelium along the embryonic axes. Development. 2006;133(12):2325–35.PubMedCrossRef
15.
Zurück zum Zitat Mailleux AA et al. Role of FGF10/FGFR2b signaling during mammary gland development in the mouse embryo. Development. 2002;129(1):53–60.PubMed Mailleux AA et al. Role of FGF10/FGFR2b signaling during mammary gland development in the mouse embryo. Development. 2002;129(1):53–60.PubMed
16.
Zurück zum Zitat Howard B et al. Identification of the scaramanga gene implicates Neuregulin3 in mammary gland specification. Genes Dev. 2005;19(17):2078–90.PubMedCrossRef Howard B et al. Identification of the scaramanga gene implicates Neuregulin3 in mammary gland specification. Genes Dev. 2005;19(17):2078–90.PubMedCrossRef
17.
Zurück zum Zitat Davenport TG, Jerome-Majewska LA, Papaioannou VE. Mammary gland, limb and yolk sac defects in mice lacking Tbx3, the gene mutated in human ulnar mammary syndrome. Development. 2003;130(10):2263–73.PubMedCrossRef Davenport TG, Jerome-Majewska LA, Papaioannou VE. Mammary gland, limb and yolk sac defects in mice lacking Tbx3, the gene mutated in human ulnar mammary syndrome. Development. 2003;130(10):2263–73.PubMedCrossRef
18.
Zurück zum Zitat Jerome-Majewska LA et al. Tbx3, the ulnar-mammary syndrome gene, and Tbx2 interact in mammary gland development through a p19Arf/p53-independent pathway. Dev Dyn. 2005;234(4):922–33.PubMedCrossRef Jerome-Majewska LA et al. Tbx3, the ulnar-mammary syndrome gene, and Tbx2 interact in mammary gland development through a p19Arf/p53-independent pathway. Dev Dyn. 2005;234(4):922–33.PubMedCrossRef
19.
Zurück zum Zitat Shermak MA. Congenital and developmental abnormalities of the breast, in Management of Breast Diseases, Jatoi I, Kaufman M, editors. 2010, Springer. p. 37–51. Shermak MA. Congenital and developmental abnormalities of the breast, in Management of Breast Diseases, Jatoi I, Kaufman M, editors. 2010, Springer. p. 37–51.
20.
Zurück zum Zitat Schmidt H. Supernumerary nipples: prevalence, size, sex and side predilection—a prospective clinical study. Eur J Pediatr. 1998;157(10):821–3.PubMedCrossRef Schmidt H. Supernumerary nipples: prevalence, size, sex and side predilection—a prospective clinical study. Eur J Pediatr. 1998;157(10):821–3.PubMedCrossRef
21.
Zurück zum Zitat Little CC, McDonald H. Abnormalities of the mammae in the house mouse. J Hered. 1945;36:285–8. Little CC, McDonald H. Abnormalities of the mammae in the house mouse. J Hered. 1945;36:285–8.
22.
Zurück zum Zitat Shiratori H, Hamada H. The left-right axis in the mouse: from origin to morphology. Development. 2006;133(11):2095–104.PubMedCrossRef Shiratori H, Hamada H. The left-right axis in the mouse: from origin to morphology. Development. 2006;133(11):2095–104.PubMedCrossRef
23.
Zurück zum Zitat Ramsdell AF. Left-right asymmetry and congenital cardiac defects: getting to the heart of the matter in vertebrate left-right axis determination. Dev Biol. 2005;288(1):1–20.PubMedCrossRef Ramsdell AF. Left-right asymmetry and congenital cardiac defects: getting to the heart of the matter in vertebrate left-right axis determination. Dev Biol. 2005;288(1):1–20.PubMedCrossRef
24.
Zurück zum Zitat Golding JP et al. Mouse myotomes pairs exhibit left-right asymmetric expression of MLC3F and alpha-skeletal actin. Dev Dyn. 2004;231(4):795–800.PubMedCrossRef Golding JP et al. Mouse myotomes pairs exhibit left-right asymmetric expression of MLC3F and alpha-skeletal actin. Dev Dyn. 2004;231(4):795–800.PubMedCrossRef
25.
Zurück zum Zitat Golding JP et al. Heparin-binding EGF-like growth factor shows transient left-right asymmetrical expression in mouse myotome pairs. Gene Expr Patterns. 2004;5(1):3–9.PubMedCrossRef Golding JP et al. Heparin-binding EGF-like growth factor shows transient left-right asymmetrical expression in mouse myotome pairs. Gene Expr Patterns. 2004;5(1):3–9.PubMedCrossRef
26.
Zurück zum Zitat Kawakami Y et al. Retinoic acid signalling links left-right asymmetric patterning and bilaterally symmetric somitogenesis in the zebrafish embryo. Nature. 2005;435(7039):165–71.PubMedCrossRef Kawakami Y et al. Retinoic acid signalling links left-right asymmetric patterning and bilaterally symmetric somitogenesis in the zebrafish embryo. Nature. 2005;435(7039):165–71.PubMedCrossRef
27.
Zurück zum Zitat Vermot J, Pourquie O. Retinoic acid coordinates somitogenesis and left-right patterning in vertebrate embryos. Nature. 2005;435(7039):215–20.PubMedCrossRef Vermot J, Pourquie O. Retinoic acid coordinates somitogenesis and left-right patterning in vertebrate embryos. Nature. 2005;435(7039):215–20.PubMedCrossRef
28.
Zurück zum Zitat Cho KW et al. Retinoic acid signaling and the initiation of mammary gland development. Dev Biol. 2012;365(1):259–66.PubMedCrossRef Cho KW et al. Retinoic acid signaling and the initiation of mammary gland development. Dev Biol. 2012;365(1):259–66.PubMedCrossRef
29.
Zurück zum Zitat Vermot J et al. Retinoic acid controls the bilateral symmetry of somite formation in the mouse embryo. Science. 2005;308(5721):563–6.PubMedCrossRef Vermot J et al. Retinoic acid controls the bilateral symmetry of somite formation in the mouse embryo. Science. 2005;308(5721):563–6.PubMedCrossRef
30.
Zurück zum Zitat Blot W, Fraumeni J, Young J. Left-sided breast cancer. Lancet. 1977;2:762–3.CrossRef Blot W, Fraumeni J, Young J. Left-sided breast cancer. Lancet. 1977;2:762–3.CrossRef
31.
Zurück zum Zitat Ekbom A et al. Epidemiologic correlates of breast cancer laterality. Cancer Causes Control. 1994;7:539–43. Ekbom A et al. Epidemiologic correlates of breast cancer laterality. Cancer Causes Control. 1994;7:539–43.
32.
Zurück zum Zitat Garinkel L, Craig L, Seidman H. An appraisal of left and right breast cancer. J Natl Cancer Inst. 1959;23:617–31. Garinkel L, Craig L, Seidman H. An appraisal of left and right breast cancer. J Natl Cancer Inst. 1959;23:617–31.
33.
Zurück zum Zitat Roychoudhuri R, Putcha V, Møller H. Cancer and laterality: a study of the five major paired organs (UK). Cancer Causes and Control. 2006;17(5):655–62.PubMedCrossRef Roychoudhuri R, Putcha V, Møller H. Cancer and laterality: a study of the five major paired organs (UK). Cancer Causes and Control. 2006;17(5):655–62.PubMedCrossRef
34.
Zurück zum Zitat Tulinius H et al. Tumors in Iceland: 10. Malignant tumours of the breast. A histological classification, laterlaity, survival and epidemiological considerations. Acta path Microbiol Immunol Scand. 1988;96:229–38. Tulinius H et al. Tumors in Iceland: 10. Malignant tumours of the breast. A histological classification, laterlaity, survival and epidemiological considerations. Acta path Microbiol Immunol Scand. 1988;96:229–38.
35.
Zurück zum Zitat Weiss H, Devesa S, Brinton L. Laterality of breast cancer. Cancer Causes Control. 1996;7:539–43.PubMedCrossRef Weiss H, Devesa S, Brinton L. Laterality of breast cancer. Cancer Causes Control. 1996;7:539–43.PubMedCrossRef
36.
Zurück zum Zitat Fatima N et al. Lower incidence but more aggressive behavior of right sided breast cancer in pakistani women: does right deserve more respect? Asian Pac J Cancer Prev. 2013;14(1):43–5.PubMedCrossRef Fatima N et al. Lower incidence but more aggressive behavior of right sided breast cancer in pakistani women: does right deserve more respect? Asian Pac J Cancer Prev. 2013;14(1):43–5.PubMedCrossRef
37.
Zurück zum Zitat Senie R et al. Epidemiology of breast carcinoma II: factors related to the predominance of left-sided disease. Cancer Causes and Control. 1980;46:1705–13. Senie R et al. Epidemiology of breast carcinoma II: factors related to the predominance of left-sided disease. Cancer Causes and Control. 1980;46:1705–13.
38.
Zurück zum Zitat Borisenkov MF and Bazhenov SM. Survival in human breast cancer: effects of tumor laterality and the time of year of surgery. Human Physiology. 2001;27(5):631-34. Borisenkov MF and Bazhenov SM. Survival in human breast cancer: effects of tumor laterality and the time of year of surgery. Human Physiology. 2001;27(5):631-34.
39.
Zurück zum Zitat Hartveit F, Tangen M, Hartveit E. Side and survival in breast cancer. Oncology. 1984;41(3):149–54.PubMedCrossRef Hartveit F, Tangen M, Hartveit E. Side and survival in breast cancer. Oncology. 1984;41(3):149–54.PubMedCrossRef
40.
Zurück zum Zitat Melnik Y et al. Breast cancer in Israel: laterality and survival. J Cancer Res Clin Oncol. 1979;95(3):291–3.PubMedCrossRef Melnik Y et al. Breast cancer in Israel: laterality and survival. J Cancer Res Clin Oncol. 1979;95(3):291–3.PubMedCrossRef
41.
Zurück zum Zitat Delahunt B, Bethwaite P, Nacey JN. Renal cell carcinoma in New Zealand: a national survival study. Urology. 1994;43(3):300–9.PubMedCrossRef Delahunt B, Bethwaite P, Nacey JN. Renal cell carcinoma in New Zealand: a national survival study. Urology. 1994;43(3):300–9.PubMedCrossRef
42.
Zurück zum Zitat Pece S, et al. Biological and molecular heterogeneity of breast cancers correlates with their cancer stem cell content. cell. 140(1): 62–73. Pece S, et al. Biological and molecular heterogeneity of breast cancers correlates with their cancer stem cell content. cell. 140(1): 62–73.
43.
Zurück zum Zitat Cappello F et al. Study of axillary lymph node asymmetry in a female population. J Anat. 2001;199(Pt 5):617–20.PubMedCrossRef Cappello F et al. Study of axillary lymph node asymmetry in a female population. J Anat. 2001;199(Pt 5):617–20.PubMedCrossRef
44.
Zurück zum Zitat Dane S et al. Asymmetries in breast cancer lateralization and both axillary lymph node number and metastatic involvement. Lymphology. 2008;41(2):75–9.PubMed Dane S et al. Asymmetries in breast cancer lateralization and both axillary lymph node number and metastatic involvement. Lymphology. 2008;41(2):75–9.PubMed
45.
Zurück zum Zitat Nouh MA et al. Lymph node metastasis in breast carcinoma: clinicopathological correlations in 3747 patients. J Egypt Natl Canc Inst. 2004;16(1):50–6.PubMed Nouh MA et al. Lymph node metastasis in breast carcinoma: clinicopathological correlations in 3747 patients. J Egypt Natl Canc Inst. 2004;16(1):50–6.PubMed
46.
Zurück zum Zitat Saleh F, Abdeen S. Pathobiological features of breast tumours in the State of Kuwait: a comprehensive analysis. J Carcinog. 2007;6:12.PubMedCrossRef Saleh F, Abdeen S. Pathobiological features of breast tumours in the State of Kuwait: a comprehensive analysis. J Carcinog. 2007;6:12.PubMedCrossRef
47.
Zurück zum Zitat Wilting J, Hagedorn M. Left-right asymmetry in embryonic development and breast cancer: common molecular determinants? Curr Med Chem. 2011;18(36):5519–27.PubMedCrossRef Wilting J, Hagedorn M. Left-right asymmetry in embryonic development and breast cancer: common molecular determinants? Curr Med Chem. 2011;18(36):5519–27.PubMedCrossRef
48.
Zurück zum Zitat Kenney NJ, Adkins HB, Sanicola M. Nodal and Cripto-1: embryonic pattern formation genes involved in mammary gland development and tumorigenesis. J Mammary Gland Biol Neoplasia. 2004;9(2):133–44.PubMedCrossRef Kenney NJ, Adkins HB, Sanicola M. Nodal and Cripto-1: embryonic pattern formation genes involved in mammary gland development and tumorigenesis. J Mammary Gland Biol Neoplasia. 2004;9(2):133–44.PubMedCrossRef
49.
Zurück zum Zitat Strizzi L et al. Emerging roles of nodal and cripto-1: from embryogenesis to breast cancer progression. Breast Dis. 2008;29(−1):91–103.PubMed Strizzi L et al. Emerging roles of nodal and cripto-1: from embryogenesis to breast cancer progression. Breast Dis. 2008;29(−1):91–103.PubMed
50.
Zurück zum Zitat Harbeck N et al. Multicenter study using paraffin-embedded tumor tissue testing PITX2 DNA methylation as a marker for outcome prediction in tamoxifen-treated, node-negative breast cancer patients. J Clin Oncol. 2008;26(31):5036–42.PubMedCrossRef Harbeck N et al. Multicenter study using paraffin-embedded tumor tissue testing PITX2 DNA methylation as a marker for outcome prediction in tamoxifen-treated, node-negative breast cancer patients. J Clin Oncol. 2008;26(31):5036–42.PubMedCrossRef
51.
Zurück zum Zitat Quail DF et al. Nodal signalling in embryogenesis and tumourigenesis. Int J Biochem Cell Biol. 2013;45(4):885–98.PubMedCrossRef Quail DF et al. Nodal signalling in embryogenesis and tumourigenesis. Int J Biochem Cell Biol. 2013;45(4):885–98.PubMedCrossRef
52.
Zurück zum Zitat Vaage J. Relationship between tumor growth characteristics and preferential sites of growth. J Natl Cancer Inst. 1984;72(5):1199–203.PubMed Vaage J. Relationship between tumor growth characteristics and preferential sites of growth. J Natl Cancer Inst. 1984;72(5):1199–203.PubMed
53.
Zurück zum Zitat Russo IH, Russo J. Hormone prevention of mammary carcinogenesis: a new approach in anticancer research. Anticancer Res. 1988;8(6 489011536):1247–64.PubMed Russo IH, Russo J. Hormone prevention of mammary carcinogenesis: a new approach in anticancer research. Anticancer Res. 1988;8(6 489011536):1247–64.PubMed
54.
Zurück zum Zitat Ball SM. The development of the terminal end bud in the prepubertal-pubertal mouse mammary gland. Anat Rec. 1998;250(4):459–64. 397175680.PubMedCrossRef Ball SM. The development of the terminal end bud in the prepubertal-pubertal mouse mammary gland. Anat Rec. 1998;250(4):459–64. 397175680.PubMedCrossRef
55.
Zurück zum Zitat Mevissen M, Haussler M, Loscher W. Alterations in ornithine decarboxylase activity in the rat mammary gland after different periods of 50 Hz magnetic field exposure. Bioelectromagnetics. 1999;20(6):338–46.PubMedCrossRef Mevissen M, Haussler M, Loscher W. Alterations in ornithine decarboxylase activity in the rat mammary gland after different periods of 50 Hz magnetic field exposure. Bioelectromagnetics. 1999;20(6):338–46.PubMedCrossRef
56.
Zurück zum Zitat Fedrowitz M, Loscher W. Exposure of Fischer 344 rats to a weak power frequency magnetic field facilitates mammary tumorigenesis in the DMBA model of breast cancer. Carcinogenesis. 2008;29(1):186–93.PubMedCrossRef Fedrowitz M, Loscher W. Exposure of Fischer 344 rats to a weak power frequency magnetic field facilitates mammary tumorigenesis in the DMBA model of breast cancer. Carcinogenesis. 2008;29(1):186–93.PubMedCrossRef
57.
Zurück zum Zitat Davie SA et al. Effects of FVB/NJ and C57Bl/6J strain backgrounds on mammary tumor phenotype in inducible nitric oxide synthase deficient mice. Transgenic Res. 2007;16(2):193–201.PubMedCrossRef Davie SA et al. Effects of FVB/NJ and C57Bl/6J strain backgrounds on mammary tumor phenotype in inducible nitric oxide synthase deficient mice. Transgenic Res. 2007;16(2):193–201.PubMedCrossRef
58.
Zurück zum Zitat Guy CT et al. Expression of the neu protooncogene in the mammary epithelium of transgenic mice induces metastatic disease. Proc Natl Acad Sci U S A. 1992;89(22):10578–82.PubMedCrossRef Guy CT et al. Expression of the neu protooncogene in the mammary epithelium of transgenic mice induces metastatic disease. Proc Natl Acad Sci U S A. 1992;89(22):10578–82.PubMedCrossRef
59.
Zurück zum Zitat Green JE et al. 2-difluoromethylornithine and dehydroepiandrosterone inhibit mammary tumor progression but not mammary or prostate tumor initiation in C3(1)/SV40 T/t-antigen transgenic mice. Cancer Res. 2001;61(20):7449–55.PubMed Green JE et al. 2-difluoromethylornithine and dehydroepiandrosterone inhibit mammary tumor progression but not mammary or prostate tumor initiation in C3(1)/SV40 T/t-antigen transgenic mice. Cancer Res. 2001;61(20):7449–55.PubMed
60.
Zurück zum Zitat Varticovski L et al. Accelerated preclinical testing using transplanted tumors from genetically engineered mouse breast cancer models. Clin Cancer Res. 2007;13(7):2168–77.PubMedCrossRef Varticovski L et al. Accelerated preclinical testing using transplanted tumors from genetically engineered mouse breast cancer models. Clin Cancer Res. 2007;13(7):2168–77.PubMedCrossRef
61.
Zurück zum Zitat Oosterhuis JW et al. The effects of regional factors on the growth rate and the differentiation of mouse teratocarcinoma. Br J Cancer. 1983;47(3):407–11. 708612944.PubMedCrossRef Oosterhuis JW et al. The effects of regional factors on the growth rate and the differentiation of mouse teratocarcinoma. Br J Cancer. 1983;47(3):407–11. 708612944.PubMedCrossRef
62.
Zurück zum Zitat Meschter CL, Connolly JM, Rose DP. Influence of regional location of the inoculation site and dietary fat on the pathology of MDA-MB-435 human breast cancer cell-derived tumors grown in nude mice. Clin Exp Metastasis. 1992;10(3 982934656):167–73.PubMedCrossRef Meschter CL, Connolly JM, Rose DP. Influence of regional location of the inoculation site and dietary fat on the pathology of MDA-MB-435 human breast cancer cell-derived tumors grown in nude mice. Clin Exp Metastasis. 1992;10(3 982934656):167–73.PubMedCrossRef
63.
Zurück zum Zitat Sheldon WG et al. Distribution of mammary gland neoplasms and factors influencing metastases in hybrid mice. Lab Anim Sci. 1982;32(2):166–8.PubMed Sheldon WG et al. Distribution of mammary gland neoplasms and factors influencing metastases in hybrid mice. Lab Anim Sci. 1982;32(2):166–8.PubMed
64.
Zurück zum Zitat Auerbach R, Auerbach W. Regional differences in the growth of normal and neoplastic cells. Science. 1982;215(4529):127–34.PubMedCrossRef Auerbach R, Auerbach W. Regional differences in the growth of normal and neoplastic cells. Science. 1982;215(4529):127–34.PubMedCrossRef
65.
Zurück zum Zitat Minasian H, Minassian D. The relationship between blood flow of mammary tissue and its overlying skin in the mouse. Lab Anim. 1983;17(4):321–3.PubMedCrossRef Minasian H, Minassian D. The relationship between blood flow of mammary tissue and its overlying skin in the mouse. Lab Anim. 1983;17(4):321–3.PubMedCrossRef
66.
Zurück zum Zitat Takebe N, Warren RQ, Ivy SP. Breast cancer growth and metastasis: interplay between cancer stem cells, embryonic signaling pathways and epithelial-to-mesenchymal transition. Breast Cancer Res. 2011;13(3):211.PubMedCrossRef Takebe N, Warren RQ, Ivy SP. Breast cancer growth and metastasis: interplay between cancer stem cells, embryonic signaling pathways and epithelial-to-mesenchymal transition. Breast Cancer Res. 2011;13(3):211.PubMedCrossRef
67.
Zurück zum Zitat Rinn JL et al. A dermal HOX transcriptional program regulates site-specific epidermal fate. Genes Dev. 2008;22(3):303–7.PubMedCrossRef Rinn JL et al. A dermal HOX transcriptional program regulates site-specific epidermal fate. Genes Dev. 2008;22(3):303–7.PubMedCrossRef
68.
Zurück zum Zitat Shah N, Sukumar S. The Hox genes and their roles in oncogenesis. Nat Rev Cancer. 2010;10(5):361–71.PubMedCrossRef Shah N, Sukumar S. The Hox genes and their roles in oncogenesis. Nat Rev Cancer. 2010;10(5):361–71.PubMedCrossRef
69.
Zurück zum Zitat Aulehla A, Pourquie O. Signaling gradients during paraxial mesoderm development. Cold Spring Harb Perspect Biol. 2010;2(2):a000869.PubMedCrossRef Aulehla A, Pourquie O. Signaling gradients during paraxial mesoderm development. Cold Spring Harb Perspect Biol. 2010;2(2):a000869.PubMedCrossRef
70.
Zurück zum Zitat Bondos S. Variations on a theme: Hox and Wnt combinatorial regulation during animal development. Sci STKE. 2006;2006(355):pe38.PubMedCrossRef Bondos S. Variations on a theme: Hox and Wnt combinatorial regulation during animal development. Sci STKE. 2006;2006(355):pe38.PubMedCrossRef
71.
Zurück zum Zitat Garcia-Gasca A, Spyropoulos DD. Differential mammary morphogenesis along the anteroposterior axis in Hoxc6 gene targeted mice. Dev Dyn. 2000;219(2):261–76.PubMedCrossRef Garcia-Gasca A, Spyropoulos DD. Differential mammary morphogenesis along the anteroposterior axis in Hoxc6 gene targeted mice. Dev Dyn. 2000;219(2):261–76.PubMedCrossRef
72.
Zurück zum Zitat Hatsell SJ, Cowin P. Gli3-mediated repression of Hedgehog targets is required for normal mammary development. Development. 2006;133(18):3661–70.PubMedCrossRef Hatsell SJ, Cowin P. Gli3-mediated repression of Hedgehog targets is required for normal mammary development. Development. 2006;133(18):3661–70.PubMedCrossRef
73.
Zurück zum Zitat Yang A et al. p63 is essential for regenerative proliferation in limb, craniofacial and epithelial development. Nature. 1999;398(6729):714–8.PubMedCrossRef Yang A et al. p63 is essential for regenerative proliferation in limb, craniofacial and epithelial development. Nature. 1999;398(6729):714–8.PubMedCrossRef
74.
Zurück zum Zitat Mills AA et al. p63 is a p53 homologue required for limb and epidermal morphogenesis. Nature. 1999;398(6729):708–13.PubMedCrossRef Mills AA et al. p63 is a p53 homologue required for limb and epidermal morphogenesis. Nature. 1999;398(6729):708–13.PubMedCrossRef
75.
Zurück zum Zitat Asselin-Labat ML et al. Gata-3 is an essential regulator of mammary-gland morphogenesis and luminal-cell differentiation. Nat Cell Biol. 2007;9(2):201–9.PubMedCrossRef Asselin-Labat ML et al. Gata-3 is an essential regulator of mammary-gland morphogenesis and luminal-cell differentiation. Nat Cell Biol. 2007;9(2):201–9.PubMedCrossRef
76.
Zurück zum Zitat Chu EY, et al. Canonical WNT signaling promotes mammary placode development and is essential for initiation of mammary gland morphogenesis. Development. 2004; 131(19): 4819–29. Chu EY, et al. Canonical WNT signaling promotes mammary placode development and is essential for initiation of mammary gland morphogenesis. Development. 2004; 131(19): 4819–29.
77.
Zurück zum Zitat Boras-Granic K et al. Lef1 is required for the transition of Wnt signaling from mesenchymal to epithelial cells in the mouse embryonic mammary gland. Dev Biol. 2006;295(1):219–31.PubMedCrossRef Boras-Granic K et al. Lef1 is required for the transition of Wnt signaling from mesenchymal to epithelial cells in the mouse embryonic mammary gland. Dev Biol. 2006;295(1):219–31.PubMedCrossRef
78.
Zurück zum Zitat Gu B et al. Pygo2 expands mammary progenitor cells by facilitating histone H3 K4 methylation. J Cell Biol. 2009;185(5):811–26.PubMedCrossRef Gu B et al. Pygo2 expands mammary progenitor cells by facilitating histone H3 K4 methylation. J Cell Biol. 2009;185(5):811–26.PubMedCrossRef
79.
Zurück zum Zitat Lindvall C et al. The Wnt signaling receptor Lrp5 is required for mammary ductal stem cell activity and Wnt1-induced tumorigenesis. J Biol Chem. 2006;281(46):35081–7.PubMedCrossRef Lindvall C et al. The Wnt signaling receptor Lrp5 is required for mammary ductal stem cell activity and Wnt1-induced tumorigenesis. J Biol Chem. 2006;281(46):35081–7.PubMedCrossRef
80.
Zurück zum Zitat Lindvall C et al. The Wnt co-receptor Lrp6 is required for normal mouse mammary gland development. PLoS One. 2009;4(6):e5813.PubMedCrossRef Lindvall C et al. The Wnt co-receptor Lrp6 is required for normal mouse mammary gland development. PLoS One. 2009;4(6):e5813.PubMedCrossRef
81.
Zurück zum Zitat Ahn Y et al. Lrp4 and Wise interplay controls the formation and patterning of mammary and other skin appendage placodes by modulating Wnt signaling. Development. 2013;140(3):583–93.PubMedCrossRef Ahn Y et al. Lrp4 and Wise interplay controls the formation and patterning of mammary and other skin appendage placodes by modulating Wnt signaling. Development. 2013;140(3):583–93.PubMedCrossRef
82.
Zurück zum Zitat Mustonen T et al. Ectodysplasin A1 promotes placodal cell fate during early morphogenesis of ectodermal appendages. Development. 2004;131(20):4907–19.PubMedCrossRef Mustonen T et al. Ectodysplasin A1 promotes placodal cell fate during early morphogenesis of ectodermal appendages. Development. 2004;131(20):4907–19.PubMedCrossRef
83.
Zurück zum Zitat Narhi K et al. Sostdc1 defines the size and number of skin appendage placodes. Dev Biol. 2012;364(2):149–61.PubMedCrossRef Narhi K et al. Sostdc1 defines the size and number of skin appendage placodes. Dev Biol. 2012;364(2):149–61.PubMedCrossRef
Metadaten
Titel
Positional Variations in Mammary Gland Development and Cancer
verfasst von
Jacqueline M. Veltmaat
Ann F. Ramsdell
Esta Sterneck
Publikationsdatum
01.06.2013
Verlag
Springer US
Erschienen in
Journal of Mammary Gland Biology and Neoplasia / Ausgabe 2/2013
Print ISSN: 1083-3021
Elektronische ISSN: 1573-7039
DOI
https://doi.org/10.1007/s10911-013-9287-3

Weitere Artikel der Ausgabe 2/2013

Journal of Mammary Gland Biology and Neoplasia 2/2013 Zur Ausgabe

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.